US20220301656A1 - Genome sequencing as an alternative to cytogenetic analysis - Google Patents

Genome sequencing as an alternative to cytogenetic analysis Download PDF

Info

Publication number
US20220301656A1
US20220301656A1 US17/699,053 US202217699053A US2022301656A1 US 20220301656 A1 US20220301656 A1 US 20220301656A1 US 202217699053 A US202217699053 A US 202217699053A US 2022301656 A1 US2022301656 A1 US 2022301656A1
Authority
US
United States
Prior art keywords
chr9
chr11
whole
chr7
genome sequencing
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/699,053
Inventor
Eric Duncavage
David Spencer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Washington University in St Louis WUSTL
Original Assignee
Washington University in St Louis WUSTL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Washington University in St Louis WUSTL filed Critical Washington University in St Louis WUSTL
Priority to US17/699,053 priority Critical patent/US20220301656A1/en
Assigned to WASHINGTON UNIVERSITY reassignment WASHINGTON UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPENCER, DAVID, DUNCAVAGE, ERIC
Publication of US20220301656A1 publication Critical patent/US20220301656A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/10Ploidy or copy number detection
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection

Definitions

  • the present disclosure generally relates to methods of genomic profiling using whole-genome sequencing.
  • Chromosome analysis has been used for cancer diagnosis and to guide treatment decisions for over 40 years.
  • the discovery of chromosome-level mutations like the BCR-ABL1 fusion gene in chronic myeloid leukemia (CML) and PML-RARA gene fusion in AML have transformed these once lethal cancers into diseases that can be essentially cured with targeted therapies.
  • CML chronic myeloid leukemia
  • PML-RARA gene fusion in AML
  • Over a thousand chromosome-level mutations have now been identified across numerous cancer types, and although more recent genomic studies of cancer have revealed many additional nucleotide-level cancer-associated mutations, cytogenetic mutations still account for the majority of clinically-relevant genomic changes in cancer.
  • FISH and karyotyping are required for the risk classification system in AML, and cytogenetic testing for chromosomal rearrangements facilitates accurate diagnosis of B-cell lymphomas and guides therapy in non-small cell lung cancer.
  • cytogenetic testing for chromosomal rearrangements facilitates accurate diagnosis of B-cell lymphomas and guides therapy in non-small cell lung cancer.
  • the oncogenes targeted by many of the FDA-approved cancer therapies result from chromosomal rearrangements that are routinely detected using karyotyping or FISH.
  • FISH testing uses locus-specific DNA probes that can miss rearrangements with atypical breakpoints or identify changes that do not result in the expected mutant gene products and may therefore not result in the anticipated clinical outcome.
  • the most limiting aspect of FISH testing is that it is impractical to test clinical samples for more than a few specific mutations at a time, which can result in testing that is incomplete if sample amounts are insufficient.
  • targeted sequencing panels e.g., the Foundation of Medicine Comprehensive Cancer Panel, some of which can detect both copy number mutations (e.g., gene deletions and amplifications) and selected gene fusion events.
  • Geno profiling is a routine component of the diagnostic workup for an increasing number of cancers and is used to predict clinical outcomes and responses to targeted therapies. Mutations that are clinically actionable for any individual type of cancer typically span a wide range of genomic events, including chromosomal rearrangements, gene amplifications and deletions, and single-nucleotide changes. The diversity of these findings necessitates the use of multiple platforms to obtain the genomic information needed for clinical management.
  • Whole-genome sequencing is an unbiased method of detecting all types of mutations that could potentially be used to replace current testing algorithms. Such sequencing can also be performed on a limited amount of DNA to identify genomic changes that may be cryptic in other types of analyses. These features of whole-genome sequencing suggest that it could improve genomic profiling in patients with cancer.
  • Genomic abnormalities are particularly important for diagnostic classification and risk assessment in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS).
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndromes
  • Recurrent chromosomal abnormalities are the basis for the AML genomic classification system of the World Health Organization, and the association of these alterations and certain genetic mutations with clinical outcomes has led to the development of algorithms for genetic risk stratification in patients with AML.
  • Similar studies involving patients with MDS have resulted in the cytogenetic component of the International Prognostic Scoring System-Revised (IPSS-R) in such patients.
  • IIPSS-R International Prognostic Scoring System-Revised
  • Fluorescence in situ hybridization (FISH) and targeted sequencing assays that use DNA, RNA, or both are also used, but these methods are informative only in the regions selected for analysis and may provide incomplete information regarding identified chromosomal rearrangements. As a result, conventional cytogenetic analysis remains an essential component of the diagnostic workup for patients with AML or MDS.
  • a computer-implemented method for the identification of clinically relevant structural variants in a subject with AML or MDS from whole genome sequencing data includes providing a whole-genome sequencing dataset, the whole-genome sequencing dataset comprising a plurality of alignments of tumor DNA sequence fragments to a reference human genome to a computing device; performing, using the computing device, a structural variant analysis on the whole-genome sequencing dataset, the structural variant analysis including copy-number alteration (CNA) identification, structural variant (SV) identification, and gene-level variant identification to identify clinically relevant structural variants indicative of AML or MDS within the whole-genome sequencing dataset; and producing, using the computing device, a report comprising the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis.
  • CNA copy-number alteration
  • SV structural variant
  • gene-level variant identification to identify clinically relevant structural variants indicative of AML or MDS within the whole-genome sequencing dataset
  • copy-number alteration (CNA) identification further comprises transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of read counts over 500,000 bp nonoverlapping windows across the genome; transforming, using the computing device, the plurality of read counts into a plurality of CNAs; and filtering, using the computing device, plurality of CNAs to retain only CNAs greater than 5 Mbp.
  • SV identification further comprises: transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of SV calls; filtering, using the computing device, the plurality of SVs to retain only SV calls greater than 100 kbp in length; and filtering, using the computing device, the SV calls greater than 100 kbp in length to identify translocations, deletions, duplications, and inversions that overlap a predefined list of recurrent and/or risk-defining SVs associated with AML or MDS.
  • gene-level variant identification further comprises identifying, using the computing device, the alignments of the whole-genome sequencing dataset within about 85 kbp targeting 40 predetermined genes and gene hotspots that are recurrently mutated in AML or MDS.
  • the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis are indicative of a clinical outcome of the subject.
  • providing the whole-genome sequencing dataset whole genome sequencing data further comprising performing whole-genome sequencing on a biological sample comprising tumor DNA from the subject with about 60 ⁇ genome coverage.
  • FIG. 1 is a block diagram schematically illustrating a system in accordance with one aspect of the disclosure.
  • FIG. 2 is a block diagram schematically illustrating a computing device in accordance with one aspect of the disclosure.
  • FIG. 3 is a block diagram schematically illustrating a remote or user computing device in accordance with one aspect of the disclosure.
  • FIG. 4 is a block diagram schematically illustrating a server system in accordance with one aspect of the disclosure.
  • FIG. 5 is a schematic illustration of the workflow and approximate processing time for each step of the rapid WGS method and analysis of samples obtained from the study patients. (An example of the reports that were generated by this process are provided in FIG. 9 ).
  • FIG. 6A is a comparison of WGS with Conventional Cytogenetic Analysis and Targeted Gene Sequencing. It shows the sensitivity of WGS for the detection of recurrent structural variants (SVs) and copy-number alterations (CNAs) as compared with conventional cytogenetic analysis and for the detection of single-nucleotide variants (SNVs) and insertion-deletions (INDELs) as compared with high-coverage targeted gene sequencing. Error bars denote 95% confidence intervals.
  • FIG. 6B shows the identification and confirmation by WGS of 13 new recurrent SVs that were not detected by conventional cytogenetic analysis, as supported by orthogonal methods, including fluorescence in situ hybridization (FISH), polymerase chain reaction (PCR) with sequencing of SV breakpoints, or detection of fusion transcripts in RNA-sequence (RNA-seq) data.
  • FISH fluorescence in situ hybridization
  • PCR polymerase chain reaction
  • RNA-seq RNA-sequence
  • FIG. 6C shows the identification of 21 new CNAs in 14 patients; 12 of these alterations were confirmed by chromosomal microarray (CMA), FISH, or sequence-defined breakpoints.
  • CMA chromosomal microarray
  • FISH chromosomal microarray
  • An additional 9 CNAs were identified by WGS only and could not be confirmed by CMA (in 6 patients) or confirmation was not attempted because of the size or abundance of the CNA event (in 3 patients).
  • CNAs were also identified in 13 patients with ambiguous or inconclusive results on cytogenetic analysis. Additional details regarding these comparisons are provided in Tables S4 and S5 and FIGS. 18A, 18B, 18C, and 18D .
  • FIG. 7A describes is a bar graph summarizing the time it took to perform WGS-Based Genomic Profiling on samples obtained from 117 consecutive patients with AML or MDS by means of WGS, as indicated by the dashed horizontal black line.
  • the height of each bar shows the total time in days for processing, starting from construction of the sequencing library and ending with completion of the automated final report for an individual patient sample.
  • the duration of each individual step (as obtained from time stamps recorded in the information management system of the clinical laboratory) is indicated by the shaded bar segments and includes the duration of library generation and quality assessment, sequencing, and analysis and reporting. These times reflect the processing time plus waiting time before the next step. Longer turnaround times occurred because of delays between steps, rather than longer processing times.
  • the dashed horizontal red lines show the recommended maximum turnaround time for FISH testing and conventional cytogenetic analysis, according to published recommendations, although shorter turnaround times occur in many laboratories.
  • FIG. 7B describes the Diagnostic Yield of WGS-based Genomic Profiling in 117 Consecutive Patients. It shows the yield of new WGS findings in samples obtained from 68 unselected, consecutive patients with AML.
  • FIG. 7B shows the cumulative number of patients with new genomic findings that were identified by WGS, as compared with conventional cytogenetic analysis or FISH, performed at the time of diagnosis, along with the cumulative number of patients with new events that changed the category of genetic risk group on the basis of established European Leukemia Network (ELN) guidelines.
  • EPN European Leukemia Network
  • FISH testing included assays for PML-RARA, CBFB-MYH11, RUNX1-RUNX1T1, del(5q), and chromosome 7 deletion, according to recommendations; all testing was performed in samples obtained from 60 of 68 patients (88%); subgroups of these assays were performed for the remaining patients.
  • the results of ELN assignments to a genetic risk group by WGS, conventional cytogenetic analysis with FISH, and cytogenetic analysis alone are shown in the top panel.
  • the red asterisk indicates that the patient's risk group was reclassified according to the WGS results, and the red arrow indicates that the risk-group assignment was based on FISH results alone.
  • FIG. 7B shows the genomic events that were detected by WGS and are labeled as concordant with cytogenetic analysis, with FISH, or with target sequencing (in black), new findings made by WGS (in blue), and new findings that resulted in a change in the ELN genetic risk group (in red).
  • the status regarding internal tandem duplication in FLT3 (FLT3-ITD) and the allele ratio as determined by PCR were used for both conventional and WGS-based risk stratifications.
  • FIG. 8A is a risk assessment by WGS in Patients with AML, according to Existing Genetic Risk Groups. It shows overall survival for 71 patients with AML who were treated with chemotherapy alone after remission, as stratified into established ELN genetic risk groups on the basis of a combination of conventional cytogenetic analysis, FISH, and targeted gene sequencing.
  • FIG. 8B shows the same cohort as in FIG. 8A with risk stratification according to WGS results.
  • FIG. 8C shows the clinical outcomes for 27 patients for whom genetic risk could not be determined because of inconclusive, unsuccessful, or unknown results on cytogenetic analysis.
  • the median survival in this cohort was 11.2 months (95% confidence interval [CI], 5.6 to 38.8).
  • FIG. 8D shows the stratification of the cohort in FIG. 8C into established genetic risk groups with the use of WGS results, which predicted shorter overall survival for patients at adverse risk than for those at intermediate or favorable risk (not adverse) (age-adjusted hazard ratio for death for intermediate or favorable risk versus adverse risk, 0.29; 95% CI, 0.09 to 0.94). All P values were calculated with the use of a log-rank test for equal survival among the groups and adjusted for multiple comparisons.
  • FIG. 9 is an exemplary cover page of a graphical ChromoSeq WGS report highlighting the clinically significant findings in the genome sequence of an AML patient.
  • FIG. 10 is a histogram distribution of genome coverage of the genome-wide coverage depth in unique reads for 235 WGS cases.
  • FIG. 11 summarizes the variants detected per patient from a number distribution of SVs, CNAs, and gene mutations detected in 235 patients.
  • FIG. 12 is series of images confirming the SVs detected using the disclosed WGSA method.
  • the images show FISH results from metaphase and interphase FISH analysis utilizing a dual color, break apart probe targeting KMT2A (manufactured by Vysis).
  • a normal signal hybridization pattern is 2Y. Rearrangement of a KMT2A locus generates a separation of the green signal, which encompasses the 5′ segment of KMT2A and surrounding region, and the red signal, which encompasses the 3′ segment of KMT2A and surrounding region.
  • FIG. 13 is a series of images showing selected CNAs obtained by FISH. See also Table S5.
  • FIG. 14A is a first normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 14B is a second normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 14C is a third normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 15A is a first normalized coverage plot of WGS results for CNAs that were detected by WGS.
  • FIG. 15B is a second normalized coverage plot of WGS results for CNAs that were detected by WGS.
  • FIG. 16 shows the sensitivity of WGS for mutations in AML risk-defining genes.
  • FIG. 17 compares FLT3-ITD detection by WGS vs. PCR. Results for 35 patients with FLT3-ITD mutations detected by either PCR and capillary electrophoresis or WGS. Rows show results for each patient with a positive ITD assay, including WGS, a clinical targeted sequencing assay, and PCR. The ITD allele sizes and allele ratios are indicated for each assay. Allele sizes and ratios were available for all positive results. Data from PCR include an in-house laboratory developed test (LDT) and the commercial FDA companion diagnostic assay, and therefore the allele size and ratios were not always reported. Note that ITD alleles were detected by WGS in two patients for whom the in-house LDT assay was negative (in 2002). ITD alleles were also detected in these patients in the AML TCGA study).
  • LDT in-house laboratory developed test
  • FIG. 18A shows the VAFs from deep targeted sequencing for 348 variants detected in 102 patients using a clinical targeted sequencing assay.
  • FIG. 18B shows the distribution of VAFs for the detected and missed variants of FIG. 18A .
  • FIG. 18C shows the abundance and coverage depth of gene mutations in WGS data for 348 variants detected in 102 patients using a clinical targeted sequencing assay.
  • Right panels show the distribution of WGS coverage for detected and missed variants.
  • FIG. 18D shows the distribution of VAFs for the detected and missed variants of FIG. 18C .
  • FIG. 19A is a coverage metric for false negative gene mutations. It shows WGS coverage (indicated by the height of the bar) and variant supporting reads (height of the blue bar) for 48 variants that were detected by clinical targeted sequencing but missed by WGS. Most of the variants (41/48) were present in the WGS data, but at very low frequency. Detection of gene variants required >3 variant-supporting reads and >6 ⁇ total coverage. Variants highlighted by the asterisk were subsequently detected upon top-up sequencing of 4 cases.
  • FIG. 19B is a coverage metric for false negative gene mutations. It demonstrates the theoretical binomial sampling probability for detecting variants at VAFs ranging from 2% to 20% and coverage levels from 0 to 100 ⁇ . Note that at 50 ⁇ coverage (the mean obtained for this study), there is a 45% probability of sampling a variant >3 times if the true abundance is 5%. This is consistent with previous work showing that ‘standard coverage’ WGS is inadequate for robust detection of low frequency variants, but this can be improved by increasing coverage depth (see also FIG. 20 ).
  • FIG. 20 shows additional variants detected after top-up sequencing.
  • FIG. 21A is the diagnostic yield from prospective sequencing of 42 consecutive MDS patients with WGS.
  • FIG. 21A is organized as in FIG. 7B .
  • Consecutive MDS patients were sequenced from April 2019 to February 2020 to estimate the diagnostic yield of WGS compared to standard testing.
  • Top panel shows the cumulative number of patients with new findings (in blue) and the cumulative number of patients with findings that changed the IPSS-R cytogenetic risk score. IPSS-R from cytogenetics and sequencing are shown below.
  • FIG. 21B shows chromosomal abnormalities obtained from WGS and indicates new findings (in blue) and findings that changed the IPSS-R category (red) in top panel.
  • Bottom panel shows mutations in MDS-associated genes referenced in NCCN guidelines, with concordance between WGS and targeted sequencing indicated by the color (concordant in black, WGS only in orange, and targeted sequencing only in green). Note that the yield in risk-defining events in this cohort is due entirely to cases where cytogenetics was unsuccessful or inconclusive, resulting in an undetermined IPSS-R risk category.
  • FIG. 22A represents AML patients with reclassified risk groups by WGS.
  • AML patients included in the outcome analysis were with defined cytogenetic risk that were reclassified by WGS. It shows the ELN risk groups by cytogenetics combined with FISH for PML-RARA, RUNX1-RUNX1T1, CBFB-MYH11, del(5q), and del(7q) (bottom), and gene mutation testing either via targeted sequencing or PCR, and WGS and FLT3-ITD PCR only (top).
  • FIG. 22B show WGS results (with the risk-defining event highlighted in red), and results from cytogenetics, FISH, and gene mutation analysis for AML patients with reclassified risk groups by WGS.
  • FLT3-ITD mutation status by PCR was used for both WGS and conventional risk group assignments.
  • Cells outlined in red indicate that testing was either not performed or failed.
  • the clinical status of each patient including whether they expired or relapsed, and the follow-up time in months from diagnosis.
  • WGS-based ELN risk group is shown in the top panel.
  • FIG. 24B are results from WGS, cytogenetics, FISH, and gene mutation testing.
  • FLT3-ITD mutation status was determined by PCR and was used for risk stratification according to ELN criteria using an allele ratio cutoff of 0.5, or presence/absence if an allele ratio was not available.
  • Bottom panel shows clinical status and follow-up time in months.
  • a rapid method based on whole-genome sequencing that recapitulates and improves upon conventional cytogenetics is disclosed.
  • Whole-genome sequencing can detect clinically relevant chromosomal rearrangements with unparalleled accuracy and would vastly improve karyotyping of tumors by making it possible to test nearly any tumor type (including formalin-fixed specimens) for virtually all clinically-relevant chromosomal rearrangements simultaneously.
  • the methods disclosed herein advance 50-year-old cytogenetic methods to the modern era by greatly expanding the range of mutations that can be detected and tumor types that can be analyzed with the ultimate goal of improving clinical decisions and patient outcomes.
  • the disclosed method includes both software and wet laboratory workflows.
  • Genetic profiling is a routine component of the diagnostic workup for an increasing number of cancers and is used to predict clinical outcomes and responses to targeted therapies. Mutations that are clinically actionable for any individual type of cancer typically span a wide range of genomic events, including chromosomal rearrangements, gene amplifications and deletions, and single-nucleotide changes. The diversity of these findings necessitates the use of multiple platforms to obtain the genetic information needed for clinical management.
  • Whole-genome sequencing is an unbiased method of detecting all types of mutations and could potentially be used to replace current testing algorithms. Such sequencing can also be performed on a limited amount of DNA and can identify genomic changes that may be cryptic in other types of analyses. These features of whole-genome sequencing make the methods disclosed herein suitable for genomic profiling in patients with cancer.
  • WGS can be performed using minimal amounts of DNA (as little as 50 ng) and from any specimen type, including formalin-fixed solid tumor tissue obtained from routine surgical biopsies, and so can be applied to nearly any tumor type and is not limited to only those tumor types with live cells. WGS procedures also do not involve complicated and time-consuming laboratory steps that are required for other sequencing methods, including complex library preparation procedures, hybridization-capture enrichment, or intact RNA, making it one of the most rapid next-generation sequencing assays and among the simplest to implement in a clinical laboratory. Finally, because WGS produces genome-wide, base-pair resolution genomic data, it can be analyzed for chromosomal rearrangements and gene-level mutations simultaneously, thereby providing a comprehensive profile of all clinically relevant mutations regardless of mutation type.
  • the disclosed method includes providing or obtaining a biological sample comprising tumor DNA.
  • a biological sample comprising tumor DNA.
  • Any suitable biological sample may be used in the disclosed method without limitation including, but not limited to, peripheral blood, bone marrow aspirate, solid tumor biopsy samples, and any other suitable biological sample.
  • the sample size may be at least about 200 ⁇ L for samples such as peripheral blood and bone marrow aspirate.
  • the disclosed method is implemented without a normal tissue comparator because the method is configured to identify clearly pathogenic somatic events that generally do not require a germline control.
  • the biological sample is provided as previously-obtained WGS data.
  • method includes extracting the tumor DNA from the biological sample using any suitable method without limitation including, but not limited to, the QIAamp DNA mini kit (Qiagen, Hilden, Germany) as detailed in the package insert, followed by quantification with the Qubit 1.0 fluorometer High Sensitivity dsDNA assay (ThermoFisher, Waltham, Mass.) as described in the Example below.
  • the method further includes performing library preparation using the tumor DNA extracted from the biological sample.
  • the amount of tumor DNA used for library preparation ranges from about 35 ng to about 500 ng or more. Any suitable method of library preparation may be used without limitation including, but not limited to the Nextera Flex library preparation kit (cat #20015804, Illumina, Inc, San Diego, Calif.) as described in the Examples below.
  • the quantified final libraries are diluted to 1 nM for equimolar pooling prior to sequencing
  • the method further includes subjecting the library to whole-genome sequencing using any suitable systems and associated methods without limitation.
  • WGS is performed using a NovaSeq 6000 sequencing instrument (Illumina) configured to obtain about 60 ⁇ genome coverage per sample.
  • the WGS may be configured to obtain a genome coverage ranging from about 10 ⁇ per sample to about 100 ⁇ per sample.
  • the WGS may be configured to obtain a genome coverage of about 10 ⁇ , 20 ⁇ , 30 ⁇ , 40 ⁇ , 50 ⁇ , 60 ⁇ , 70 ⁇ , 80 ⁇ , 90 ⁇ , 100 ⁇ , or more per sample.
  • the method further includes aligning the WGS data to a human reference genome including, but not limited to the GRCh38 human reference genome.
  • Any suitable alignment method may be used without limitation including local alignment software such as DRAGEN (version 3.5.7) hardware-accelerated sequence processing software suite or cloud-based alignment software such as the DRAGEN Germline (alignment only) BaseSpace App.
  • the alignments are provided for variant analysis in any suitable format including, but not limited to CRAM format.
  • the disclosed method makes use of a streamlined approach for rapid whole-genome sequencing and analysis that detects clinically relevant cytogenetic abnormalities in a range of tumor types.
  • the ChromoSeq WGS assay is a high-performance tumor-only WGS analysis pipeline that generates a digital karyotype and detects known chromosomal rearrangements and gene-level mutations based on analysis of raw WGS sequence data produced from a single biological sample.
  • the method further includes performing variant analysis on the alignments using the ChromoSeq WGS assay.
  • Variant analysis includes CNA identification, SV identification, and gene-level variant identification.
  • each portion of the variant analysis is limited to targeted analysis and filtering to streamline the process while yielding variants that are be clinically relevant.
  • each portion of the variant analysis is performed using the same alignments from the single biological sample.
  • the alignments are transformed into read counts in 500,000 bp nonoverlapping windows across the genome and the read counts are transformed into CNAs using any suitable method including, but not limited to, the purity and subclone-aware Hidden Markov Model, ichorCNA as described in the examples below.
  • the reported CNAs are filtered to remove CNAs ⁇ 5 Mbp; cytogenetically evident CNAs are typically greater than plurality of CNAs, a size potentially detectable by karyotype analysis.
  • a break-end caller Manta is used to detect SV calls of at least 100 kbp in length to reduce the number of calls with unknown clinical significance.
  • the detected SVs are then filtered to identify translocations, deletions, duplications, and inversions that overlap a curated list of 612 recurrent and/or risk-defining SVs obtained from published sources, including the WHO and the Atlas of Genetics and Cytogenetics in Oncology.
  • a list of the recurrent and/or risk-defining SVs is provided as Table S2 in the examples below. Genomic events where both ends overlap one of these recurrent SVs are reported as ‘top-level’ findings in ChromoSeq without additional filtering.
  • the filtering criteria may include retaining those SVs based on: 1) at least 2 ‘paired and 2 ‘split’ reads supporting the break-ends, 2) absence of an overlapping call from a large set of SVs identified from >17,795 human genomes, 3) coverage depth of deletion or duplication call must be ⁇ 0.8 or >1.3 compared to the background, respectively, and 4) a defined breakpoint must be identified and the spanning contig generated by Manta must map back to the reported breakpoints.
  • Gene-level variants are identified using the same alignments from the biological sample as used for CNA and SV analysis.
  • gene mutations are identified within about 85 kbp targeting 40 genes and gene hotspots that are recurrently mutated in AML or MDS.
  • an indel caller is run on exons 13-15 of FLT3 to identify FLT3 ITD alleles.
  • the method includes combining the annotated CNA, SV, and gene mutation calls obtained as described above are combined with coverage QC information to generate a final text report as well as data files (CRAM, and VCF) and graphical coverage plots from ichorCNA.
  • a graphical report is generated, as shown in FIG. 9 .
  • the disclosed method detects clinically significant structural variants (copy number alterations and translocations) from WGS data, and provides the ability to replicate and outperform conventional cytogenetic testing.
  • the ChromoSeq WGS automated workflow sifts down up to 10,000 or more potential CAN and SV calls to 1 or 2 real events without sequencing paired normal tissue, greatly streamlining and simplifying WGS for use as a clinical assay.
  • the disclosed methods are streamlined at each phase to provide for clinically timely results.
  • scalable methods of sample preparation that can be performed by a single technician in less than 8 hours with commercially available reagents are used.
  • the samples are subjected to high-throughput sequencing followed by automated tumor-only variant analysis to detect mutations in selected genes, copy-number alterations of more than 5 Mbp, and recurrent structural variants.
  • the method includes automatically generating the findings of the analysis in a concise clinical report.
  • the methods disclosed herein obviate at least a portion of the challenges associated with the use of WGS for clinical testing, in particular the high cost of sequencing and the complex analysis of results that are typically too complicated and time consuming for clinical laboratories.
  • the disclosed methods include a simplified, tumor-only WGS analysis strategy that focuses on detecting known, clinically-relevant chromosome-level mutations that are routinely tested for by clinical cytogenetic laboratories.
  • the disclosed method greatly reduces the time, cost, and complexity of WGS, while also expanding the number of mutations that can be queried in each sample. Previous research of tumors using WGS have been focused on comprehensive discovery of new mutations, rather than efficient and comprehensive detection of known mutations.
  • the disclosed method makes use of recently-developed sequencing systems including, but not limited to, the Illumina NovaSeq 6000 instrument to perform WGS of tumor samples.
  • Such systems are capable of generating high coverage WGS data in a matter of days at a cost that is comparable to standard karyotyping and cytogenetic analysis that is typically used for clinical testing.
  • a streamlined approach to whole-genome sequencing is disclosed.
  • the disclosed method is designed to provide comprehensive genomic profiling of clinically relevant mutations in samples obtained from patients with AML or MDS, while minimizing the turnaround time and technical complexity.
  • An overview of the disclosed method is provided in FIG. 5 .
  • scalable methods of sample preparation that can be performed by a single technician in less than 8 hours with commercially available reagents were used, followed by standard high-throughput sequencing.
  • Automated tumor-only variant analysis detected mutations in selected genes, copy-number alterations of more than 5 Mbp, and recurrent structural variants (Tables S1 and S2).
  • the findings of the WGS analysis is summarized in a concise clinical report to a practitioner ( FIG. 9 ).
  • Genomic abnormalities are particularly important for diagnostic classification and risk assessment in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS).
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndromes
  • Recurrent chromosomal abnormalities are the basis for the AML genomic classification system of the World Health Organization, and the association of these alterations and certain genetic mutations with clinical outcomes has led to the development of algorithms for genetic risk stratification in patients with AML.
  • Similar studies involving patients with MDS have resulted in the cytogenetic component of the International Prognostic Scoring System-Revised (IPSS-R) in such patients.
  • IIPSS-R International Prognostic Scoring System-Revised
  • FISH Fluorescence in situ hybridization
  • targeted sequencing assays that use DNA, RNA, or both are also used, but these methods are informative only in the regions selected for analysis and may provide incomplete information regarding identified chromosomal rearrangements.
  • conventional cytogenetic analysis remains an essential component of the diagnostic workup for patients with AML or MDS.
  • the method includes the use of whole-genome sequencing in place of standard testing for genetic profiling in AML and MDS patients.
  • whole-genome sequencing in place of standard testing for genetic profiling in AML and MDS patients.
  • the diagnostic yield of whole-genome sequencing will depend on laboratory-specific karyotyping practices and the use of FISH or other ancillary testing; and some rapid diagnostic assays may still be used for urgent treatment decisions (e.g., FISH or quantitative PCR for PML-RARA rearrangements and PCR for FLT3-ITD mutations).
  • FISH or quantitative PCR for PML-RARA rearrangements and PCR for FLT3-ITD mutations e.g., FISH or quantitative PCR for PML-RARA rearrangements and PCR for FLT3-ITD mutations.
  • the Examples below demonstrate that whole-genome sequencing provides definitive results for clinically relevant genomic events with the use of a single test.
  • WGS results are suitable for use in risk predictions with existing, clinically validated risk-stratification systems.
  • the disclosed method adds prognostic value by expanding risk stratification to more patients, especially for those with inconclusive results on cytogenetic analysis, where whole-genome sequencing could have an immediate effect on treatment decisions.
  • Whole-genome sequencing can be performed on DNA from tissue biopsy samples of solid tumors, which are often insufficient for standard molecular assays and difficult to culture for cytogenetic studies.
  • the benefits of WGS may be even greater for these cancer types, in which whole-genome sequencing could be used to rapidly survey the entire genome for an expanding number of key mutations and structural alterations with only a small amount of DNA.
  • Such an approach would simplify genomic testing for these patients and probably increase the yield of clinically relevant findings, which improve the precision of approaches for treating many patients with cancer.
  • At least a portion of the disclosed whole-genome sequencing methods may be implemented using various computing systems and devices as described below.
  • FIG. 1 depicts a simplified block diagram of a computing device for implementing the methods described herein.
  • the computing device 300 may be configured to implement at least a portion of the tasks associated with the disclosed method using a whole-genome sequencing system 310 including, but not limited to: operating the sequencing system 310 to obtain whole-genome sequencing (WGS) data, analyzing the WGS data to identify mutations, copy-number alterations, structural variants, and generating a clinical report of findings.
  • the computer system 300 may include a computing device 302 .
  • the computing device 302 is part of a server system 304 , which also includes a database server 306 .
  • the computing device 302 is in communication with a database 308 through the database server 306 .
  • the computing device 302 is communicably coupled to the sequencing system 310 and a user-computing device 330 through a network 350 .
  • the network 350 may be any network that allows local area or wide area communication between the devices.
  • the network 350 may allow communicative coupling to the Internet through at least one of many interfaces including, but not limited to, at least one of a network, such as the Internet, a local area network (LAN), a wide area network (WAN), an integrated services digital network (ISDN), a dial-up-connection, a digital subscriber line (DSL), a cellular phone connection, and a cable modem.
  • LAN local area network
  • WAN wide area network
  • ISDN integrated services digital network
  • DSL digital subscriber line
  • cellular phone connection and a cable modem.
  • the user-computing device 330 may be any device capable of accessing the Internet including, but not limited to, a desktop computer, a laptop computer, a personal digital assistant (PDA), a cellular phone, a smartphone, a tablet, a phablet, wearable electronics, smartwatch, or other web-based connectable equipment or mobile devices.
  • a desktop computer a laptop computer
  • PDA personal digital assistant
  • a cellular phone a smartphone
  • a tablet a phablet
  • wearable electronics smartwatch
  • smartwatch or other web-based connectable equipment or mobile devices.
  • FIG. 2 depicts a component configuration 400 of computing device 402 , which includes database 410 along with other related computing components.
  • computing device 402 is similar to computing device 302 (shown in FIG. 1 ).
  • a user 404 may access components of computing device 402 .
  • database 410 is similar to database 308 (shown in FIG. 1 ).
  • database 410 includes sequencing data 418 and algorithm data 420 .
  • suitable sequencing data 418 include any data associated with the whole genome sequencing and alignment.
  • suitable algorithm data 420 include any values of parameters defining the analysis of the whole genome sequencing data, such as any of the parameters defining the WGS library, variant analysis, copy-number alteration identification, and structural variant analysis.
  • Other non-limiting examples of suitable algorithm data 420 include any parameters defining the formatting of a clinical report of results.
  • Computing device 402 also includes a number of components that perform specific tasks.
  • computing device 402 includes a data storage device 430 , segmentation component 440 , analysis component 450 , and communication component 460 .
  • Data storage device 430 is configured to store data received or generated by computing device 402 , such as any of the data stored in database 410 or any outputs of processes implemented by any component of computing device 402 .
  • Sequencing component 440 is configured to operate or produce signals configured to operate, a sequencing system to obtain and align whole-genome sequencing data.
  • Analysis component 450 is configured to analyze the WGS data and generate clinical reports as described herein.
  • Communication component 460 is configured to enable communications between computing device 402 and other devices (e.g. user computing device 330 and sequencing system 310 , shown in FIG. 1 ) over a network, such as network 350 (shown in FIG. 1 ), or a plurality of network connections using predefined network protocols such as TCP/IP (Transmission Control Protocol/Internet Protocol).
  • a network such as network 350 (shown in FIG. 1 ), or a plurality of network connections using predefined network protocols such as TCP/IP (Transmission Control Protocol/Internet Protocol).
  • TCP/IP Transmission Control Protocol/Internet Protocol
  • FIG. 3 depicts a configuration of a remote or user-computing device 502 , such as user computing device 330 (shown in FIG. 1 ).
  • Computing device 502 may include a processor 505 for executing instructions.
  • executable instructions may be stored in a memory area 510 .
  • Processor 505 may include one or more processing units (e.g., in a multi-core configuration).
  • Memory area 510 may be any device allowing information such as executable instructions and/or other data to be stored and retrieved.
  • Memory area 510 may include one or more computer-readable media.
  • Computing device 502 may also include at least one media output component 515 for presenting information to a user 501 .
  • Media output component 515 may be any component capable of conveying information to user 501 .
  • media output component 515 may include an output adapter, such as a video adapter and/or an audio adapter.
  • An output adapter may be operatively coupled to processor 505 and operatively coupleable to an output device such as a display device (e.g., a liquid crystal display (LCD), organic light emitting diode (OLED) display, cathode ray tube (CRT), or “electronic ink” display) or an audio output device (e.g., a speaker or headphones).
  • a display device e.g., a liquid crystal display (LCD), organic light emitting diode (OLED) display, cathode ray tube (CRT), or “electronic ink” display
  • an audio output device e.g., a speaker or headphones.
  • computing device 502 may include an input device 520 for receiving input from user 501 .
  • Input device 520 may include, for example, a keyboard, a pointing device, a mouse, a stylus, a touch-sensitive panel (e.g., a touchpad or a touch screen), a camera, a gyroscope, an accelerometer, a position detector, and/or an audio input device.
  • a single component such as a touch screen may function as both an output device of media output component 515 and input device 520 .
  • Computing device 502 may also include a communication interface 525 , which may be communicatively coupleable to a remote device.
  • Communication interface 525 may include, for example, a wired or wireless network adapter or a wireless data transceiver for use with a mobile phone network (e.g., Global System for Mobile communications (GSM), 3G, 4G or Bluetooth) or other mobile data network (e.g., Worldwide Interoperability for Microwave Access (WIMAX)).
  • GSM Global System for Mobile communications
  • 3G, 4G or Bluetooth or other mobile data network
  • WIMAX Worldwide Interoperability for Microwave Access
  • Stored in memory area 510 are, for example, computer-readable instructions for providing a user interface to user 501 via media output component 515 and, optionally, receiving and processing input from input device 520 .
  • a user interface may include, among other possibilities, a web browser and client application. Web browsers enable users 501 to display and interact with media and other information typically embedded on a web page or a website from a web server.
  • a client application allows users 501 to interact with a server application associated with, for example, a vendor or business.
  • FIG. 4 illustrates an example configuration of a server system 602 .
  • Server system 602 may include, but is not limited to, database server 306 and computing device 302 (both shown in FIG. 1 ). In some aspects, server system 602 is similar to server system 304 (shown in FIG. 1 ).
  • Server system 602 may include a processor 605 for executing instructions. Instructions may be stored in a memory area 625 , for example.
  • Processor 605 may include one or more processing units (e.g., in a multi-core configuration).
  • Processor 605 may be operatively coupled to a communication interface 615 such that server system 602 may be capable of communicating with a remote device such as user computing device 330 (shown in FIG. 1 ) or another server system 602 .
  • communication interface 615 may receive requests from user computing device 330 via a network 350 (shown in FIG. 1 ).
  • Storage device 625 may be any computer-operated hardware suitable for storing and/or retrieving data.
  • storage device 625 may be integrated in server system 602 .
  • server system 602 may include one or more hard disk drives as storage device 625 .
  • storage device 625 may be external to server system 602 and may be accessed by a plurality of server systems 602 .
  • storage device 625 may include multiple storage units such as hard disks or solid-state disks in a redundant array of inexpensive disks (RAID) configuration.
  • Storage device 625 may include a storage area network (SAN) and/or a network attached storage (NAS) system.
  • SAN storage area network
  • NAS network attached storage
  • processor 605 may be operatively coupled to storage device 625 via a storage interface 620 .
  • Storage interface 620 may be any component capable of providing processor 605 with access to storage device 625 .
  • Storage interface 620 may include, for example, an Advanced Technology Attachment (ATA) adapter, a Serial ATA (SATA) adapter, a Small Computer System Interface (SCSI) adapter, a RAID controller, a SAN adapter, a network adapter, and/or any component providing processor 605 with access to storage device 625 .
  • ATA Advanced Technology Attachment
  • SATA Serial ATA
  • SCSI Small Computer System Interface
  • Memory areas 510 (shown in FIG. 3 ) and 610 may include, but are not limited to, random access memory (RAM) such as dynamic RAM (DRAM) or static RAM (SRAM), read-only memory (ROM), erasable programmable read-only memory (EPROM), electrically erasable programmable read-only memory (EEPROM), and non-volatile RAM (NVRAM).
  • RAM random access memory
  • DRAM dynamic RAM
  • SRAM static RAM
  • ROM read-only memory
  • EPROM erasable programmable read-only memory
  • EEPROM electrically erasable programmable read-only memory
  • NVRAM non-volatile RAM
  • the computer systems and computer-implemented methods discussed herein may include additional, less, or alternate actions and/or functionalities, including those discussed elsewhere herein.
  • the computer systems may include or be implemented via computer-executable instructions stored on non-transitory computer-readable media.
  • the methods may be implemented via one or more local or remote processors, transceivers, servers, and/or sensors (such as processors, transceivers, servers, and/or sensors mounted on vehicle or mobile devices, or associated with smart infrastructure or remote servers), and/or via computer executable instructions stored on non-transitory computer-readable media or medium.
  • a computing device is configured to implement machine learning, such that the computing device “learns” to analyze, organize, and/or process data without being explicitly programmed.
  • Machine learning may be implemented through machine learning (ML) methods and algorithms.
  • a machine learning (ML) module is configured to implement ML methods and algorithms.
  • ML methods and algorithms are applied to data inputs and generate machine learning (ML) outputs.
  • Data inputs may further include: sequencing data, sensor data, image data, video data, telematics data, authentication data, authorization data, security data, mobile device data, geolocation information, transaction data, personal identification data, financial data, usage data, weather pattern data, “big data” sets, and/or user preference data.
  • data inputs may include certain ML outputs.
  • At least one of a plurality of ML methods and algorithms may be applied, which may include but are not limited to: linear or logistic regression, instance-based algorithms, regularization algorithms, decision trees, Bayesian networks, cluster analysis, association rule learning, artificial neural networks, deep learning, dimensionality reduction, and support vector machines.
  • the implemented ML methods and algorithms are directed toward at least one of a plurality of categorizations of machine learning, such as supervised learning, unsupervised learning, and reinforcement learning.
  • ML methods and algorithms are directed toward supervised learning, which involves identifying patterns in existing data to make predictions about subsequently received data.
  • ML methods and algorithms directed toward supervised learning are “trained” through training data, which includes example inputs and associated example outputs.
  • the ML methods and algorithms may generate a predictive function that maps outputs to inputs and utilize the predictive function to generate ML outputs based on data inputs.
  • the example inputs and example outputs of the training data may include any of the data inputs or ML outputs described above.
  • ML methods and algorithms are directed toward unsupervised learning, which involves finding meaningful relationships in unorganized data. Unlike supervised learning, unsupervised learning does not involve user-initiated training based on example inputs with associated outputs. Rather, in unsupervised learning, unlabeled data, which may be any combination of data inputs and/or ML outputs as described above, is organized according to an algorithm-determined relationship.
  • ML methods and algorithms are directed toward reinforcement learning, which involves optimizing outputs based on feedback from a reward signal.
  • ML methods and algorithms directed toward reinforcement learning may receive a user-defined reward signal definition, receive a data input, utilize a decision-making model to generate an ML output based on the data input, receive a reward signal based on the reward signal definition and the ML output, and alter the decision-making model so as to receive a stronger reward signal for subsequently generated ML outputs.
  • the reward signal definition may be based on any of the data inputs or ML outputs described above.
  • an ML module implements reinforcement learning in a user recommendation application.
  • the ML module may utilize a decision-making model to generate a ranked list of options based on user information received from the user and may further receive selection data based on a user selection of one of the ranked options.
  • a reward signal may be generated based on comparing the selection data to the ranking of the selected option.
  • the ML module may update the decision-making model such that subsequently generated rankings more accurately predict a user selection.
  • any such resulting program, having computer-readable code means may be embodied or provided within one or more computer-readable media, thereby making a computer program product, i.e., an article of manufacture, according to the discussed aspects of the disclosure.
  • the computer-readable media may be, for example, but is not limited to, a fixed (hard) drive, diskette, optical disk, magnetic tape, semiconductor memory such as read-only memory (ROM), and/or any transmitting/receiving medium, such as the Internet or other communication network or link.
  • the article of manufacture containing the computer code may be made and/or used by executing the code directly from one medium, by copying the code from one medium to another medium, or by transmitting the code over a network.
  • a processor may include any programmable system including systems using micro-controllers, reduced instruction set circuits (RISC), application specific integrated circuits (ASICs), logic circuits, and any other circuit or processor capable of executing the functions described herein.
  • RISC reduced instruction set circuits
  • ASICs application specific integrated circuits
  • logic circuits and any other circuit or processor capable of executing the functions described herein.
  • the above examples are examples only, and are thus not intended to limit in any way the definition and/or meaning of the term “processor.”
  • the terms “software” and “firmware” are interchangeable, and include any computer program stored in memory for execution by a processor, including RAM memory, ROM memory, EPROM memory, EEPROM memory, and non-volatile RAM (NVRAM) memory.
  • RAM random access memory
  • ROM memory read-only memory
  • EPROM memory erasable programmable read-only memory
  • EEPROM memory electrically erasable programmable read-only memory
  • NVRAM non-volatile RAM
  • a computer program is provided, and the program is embodied on a computer-readable medium.
  • the system is executed on a single computer system, without requiring a connection to a server computer.
  • the system is being run in a Windows® environment (Windows is a registered trademark of Microsoft Corporation, Redmond, Wash.).
  • the system is run on a mainframe environment and a UNIX® server environment (UNIX is a registered trademark of X/Open Company Limited located in Reading, Berkshire, United Kingdom).
  • the application is flexible and designed to run in various different environments without compromising any major functionality.
  • the system includes multiple components distributed among a plurality of computing devices.
  • One or more components may be in the form of computer-executable instructions embodied in a computer-readable medium.
  • the systems and processes are not limited to the specific aspects described herein.
  • components of each system and each process can be practiced independent and separate from other components and processes described herein.
  • Each component and process can also be used in combination with other assembly packages and processes.
  • the present aspects may enhance the functionality and functioning of computers and/or computer systems.
  • numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth, used to describe and claim certain embodiments of the present disclosure are to be understood as being modified in some instances by the term “about.”
  • the term “about” is used to indicate that a value includes the standard deviation of the mean for the device or method being employed to determine the value.
  • the numerical parameters set forth in the written description and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment.
  • the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
  • the terms “a” and “an” and “the” and similar references used in the context of describing a particular embodiment (especially in the context of certain of the following claims) can be construed to cover both the singular and the plural, unless specifically noted otherwise.
  • the term “or” as used herein, including the claims, is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive.
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndromes
  • 111 samples were selected based on DNA availability and results from conventional cytogenetic studies in order to include a wide range of chromosomal abnormalities, including risk-defining translocations, copy number alterations (CNAs), and either a complex or normal karyotype.
  • CNAs copy number alterations
  • samples with successful cytogenetic studies from the prospective cohort below were also used for WGS performance evaluation, and likewise, prospective samples with unsuccessful or inconclusive cytogenetics were used for the analysis of WGS-based risk prediction in patients with unknown cytogenetics.
  • Tumor-only WGS was performed in a CLIA-licensed environment clinical sequencing laboratory; no normal tissue comparator was used for this assay in order to reduce time, complexity, and cost, and because the purpose is to identify clearly pathogenic somatic events. that generally do not require a germline control. All samples were accessioned into the MGI laboratory information management system (LIMS) upon receipt prior to DNA extraction (for prospective samples) or library preparation (for retrospective samples received as DNA).
  • LIMS MGI laboratory information management system
  • DNA from prospective peripheral blood or bone marrow aspirate specimens was extracted using 200 uL of material with the QIAamp DNA mini kit (Qiagen, Hilden, Germany) as detailed in the package insert, followed by quantification with the Qubit 1.0 fluorometer High Sensitivity dsDNA assay (ThermoFisher, Waltham, Mass.). Subsequent WGS procedures are described below.
  • WGS library preparation used the Nextera Flex library preparation kit (cat #20015804, Illumina, Inc, San Diego, Calif.) along with dual unique index library adapters (cat #20015881).
  • This onbead tagmentation-based library construction method was selected because it is fast, simple, and automatable, and thus fits well in a clinical testing environment where training of laboratory staff and turnaround time are important considerations.
  • library construction was performed in a single day in batches of 2 to 16 samples by individual laboratory staff who followed the protocol detailed in the package insert without modification. In general, 500 ng of input DNA was used for library construction, although as little as 35 ng was used when DNA amounts were limiting.
  • Sequencing was performed on NovaSeq 6000 sequencing instruments (Illumina) using either S1 or S4 flowcells and 2 ⁇ 150 sequencing chemistry. Retrospective samples were sequenced on S4 flowcells in pools of 16 (or pools of 4 samples on one S4 lane using the XP lane loader) and prospective ‘real-time’ sequencing used 51 flow cells in pools of 3 samples, which is designed to yield >133 Gbp of raw sequence and 60 ⁇ genome coverage per sample. Flowcell loading and sequencing were performed as recommended by the manufacturer. Times for sequencing are 25 and 44 hours for 51 and S4 flowcells, respectively, and were documented in the MGI LIMS system.
  • Tumor-only variant analysis used a custom analysis workflow (‘ChromoSeq’) specified in the WDL workflow language and executed using the Cromwell workflow engine1 in dockerized compute containers, which is available for public use as a custom application on BaseSpace (https://basespace.illumina.com/apps/6984978/Chromoseq—pending BaseSpace approval for public release).
  • Analysis involves three components: CNA identification, SV identification, and gene-level variant identification. All three of these components are subject to targeted analysis and filtering to yield variants that may be clinically relevant.
  • Cytogenetically evident CNAs greater than 5 Mbp which are of a size potentially detectable by karyotype analysis, are identified via a read-depth approach using a previously published purity and subclone-aware Hidden Markov Model, ichorCNA2; https://github.com/GavinHaLab/ichorCNA).
  • the input for this script is a file with read counts in 500,000 bp nonoverlapping windows across the genome, either generated using bedtools3 or outputted directly from the DRAGEN mapping software during alignment using the command: dragen -r Sobj-> ⁇ dragenref ⁇ --fastq-list $fastqfile --fastq-list-sample-id $samplename --enable-cnv true --cnv-targetbed/staging/garza testing/reference/all sequences.fa.bed --cnv-interval-width 500000 --output-directory $cramout output-file-prefix $sample --output-format CRAM --enable-bam-indexing true --enable-duplicate-marking true.
  • Binned read counts are supplied to ichorCNA for normalization for GC content, mappability, and using a ‘panel of normals’ normalization file generated from 20 normal karyotype cases per the instructions on the ichorCNA github repo.
  • Outputs from ichorCNA were text-processed via a custom PERL script (available upon request) to retain CNAs >5 Mbp and converted to VCF format, and then combined with SV calls (below) for input into the ChromoSeq reporting script.
  • L2R is the log 2 normalized coverage ratio vs. a panel of normals and CN is the estimated copy number for the event.
  • SV identification is performed with the break-end caller Manta and broken into two ‘tiers’.
  • first tier recurrrent and risk defining events are detected with a high sensitivity approach
  • second tier novel SVs are subject to more rigorous filtering.
  • Manta is run directly from the aligned CRAM file in ‘tumor’ mode and with custom parameters to increase the sensitivity and limit calls to those that are at least 100 kbp in length to reduce the number of calls with unknown clinical significance.
  • SVs are then filtered to identify translocations, deletions, duplications, and inversions that overlap a curated list of 612 recurrent and/or risk-defining SVs obtained from published sources, including the WHO and the Atlas of Genetics and Cytogenetics in Oncology (see Table S2). Genomic events where both ends overlap one of these recurrent SVs are reported as ‘top-level’ findings in ChromoSeq without additional filtering. Although the remaining SVs will rarely be clinically relevant, they could include patient-specific events or identify cytogenetically cryptic rearrangements involving genes relevant for AML or MDS. We therefore perform rigorous annotation and filtering using a custom PERL script (available upon request) and report the remaining high-quality novel events as secondary findings.
  • Gene mutations are identified in—85 kbp targeting 40 genes and gene hotspots that are recurrently mutated in AML or MDS8. This target space was selected to be identical to that of the targeted gene panel used for clinical testing of these patients at our institution, and is relatively small so that rare inherited variants (i.e., variants of uncertain significance (VUS)), are minimized.
  • the primary variant caller is Varscan2, which is run in SNV and indel mode using custom parameters to enhance sensitivity.
  • the indel caller Pindel and Manta are also run on exons 13-15 of FLT3 to identify FLT3 ITD alleles.
  • a read count based ‘hotspot’ analysis is performed on 66 recurrently mutated positions to recover low abundance variants that are not detected by Varscan2 (a minimum variant read count of 3 is required to report these hotspot positions).
  • Variant calls identified via these approaches are merged and harmonized using a custom python script (available upon request), and annotated with VEP using Ensembl version 90 prior to reporting.
  • Annotated CNA, SV, and gene mutation calls are combined with coverage QC information to generate a final text report using a custom python script (available upon request).
  • This report includes the CNAs, recurrent SVs, and gene mutations identified by the above steps as ‘toplevel’ results.
  • the remaining SVs that remained after filtering are reported in two categories. The first includes high-quality novel SVs that affect (overlap) a gene that is included in either the recurrent SV or gene mutation target space.
  • the second category is all other high-quality novel SVs. Additional coverage QC metrics are also reported.
  • This text file is copied to the final case directory along with data files (CRAM, and VCF) and graphical coverage plots from ichorCNA.
  • the final text report is also used to generate a graphical ChromoSeq report, as shown in FIG. 9 .
  • ChromoSeq reports with QC metrics and variants for the prospective patients were reviewed in 1 hour sessions by board-certified molecular pathologists and a board-certified cytogeneticist and molecular geneticist without prior knowledge of the results from conventional testing. Exact times for processing steps shown in FIG. 7A were obtained from the MGI LIMS system and from the timestamp in the ChromoSeq text report. A final ChromoSeq analysis was performed on all prospective samples at the end of the study to harmonize the results and file formats (which changed over the course of the study) with the outputs from the retrospective samples.
  • cytogenetic and FISH analyses were performed according to standard clinical protocols. We obtained data regarding genetic mutations as part of standard diagnostic testing using polymerase-chain-reaction (PCR)—based assays for the internal tandem duplication mutation in FLT3 (FLT3-ITD) and the NPM1c mutation, a laboratory-developed clinical sequencing assay, or both. Cytogenetic and molecular results were used to assign patients to established European Leukemia Network (ELN) or IPSS-R risk categories.
  • EPN European Leukemia Network
  • IPSS-R risk categories IPSS-R risk categories.
  • Gene mutations were obtained as part of standard diagnostic testing using a commercially available PCR-based assay for the FLT3 internal tandem duplication mutation (ITD) (Invivoscribe, San Diego, Calif.), in-house testing for the NPM1c mutation, and/or a laboratory-developed clinical sequencing assay, including either clinical tumor/normal exome sequencing or a clinical gene panel that targets 40 recurrently mutated genes or gene hotspots in AML and MDS (Myeloseq; Department of Pathology and Immunology, Washington University School of Medicine, see Table
  • WGS results were compared to conventional cytogenetics and FISH to determine the sensitivity and positive predictive value for detecting recurrent SVs and CNAs. These comparisons used the following approaches:
  • FISH was the primary means of confirmation for new SVs and CNAs when appropriate probes were available and clinical specimens were available for testing. All FISH studies were performed using validated probes and standard clinical procedures using 200 cells and were reviewed by board certified cytogeneticists. The presence of an abnormal result in the specified study was considered as support for the genomic event identified by WGS. For example, we considered an abnormal result for the KMT2A dual color/dual fusion FISH assay as confirmation of an SV involving KTM2A in the WGS data.
  • PCR primers were designed from breakpoint-spanning sequence contigs generated from Manta and were used in standard PCR reactions with human genomic DNA. Amplified fragments were excised, purified, sequenced with Sanger sequencing, and analyzed with Blat to verify localization to the breakpoint region.
  • CNAs were confirmed via chromosomal microarray (CMA) for cases with available DNA but insufficient material or probe for FISH assays.
  • CMAs were performed per standard methods using the CytoScan HD platform (ThermoFisher) with subsequent analysis in Chromosome Analysis Suite (ThermoFisher). Data were reviewed and interpreted by a board-certified cytogeneticist and molecular geneticist.
  • SVs in two cases with KMT2A rearrangements were confirmed using existing RNA-seq data that was published as part of the TCGA AML study (see Supplemental Table 1 in ref 18, which can be accessed here: https://api.gdc.cancer.gov/data/b9196563-a05d-40b8-80dc-640ec712eb06; samples 380949 and 410324).
  • Clinical FISH using a KMT2A breakapart probe for these cases was also abnormal, and the identification of a fusion transcript via RNA-seq provided the definitive confirmation of the translocation partner.
  • T provide a basis of comparison for risk stratification results obtained using the disclosed WGS method, cytogenetics, FISH, and molecular results were used to assign patients to established genomic risk categories, which used the 2017 ELN guidelines for AML patients12 and the cytogenetic component of the IPSS-R scoring system for MDS patients, both without modification. Cytogenetic abnormalities were required to meet the abovementioned criteria to be considered clonal.
  • IPSS-R risk groups do not involve gene mutations and are therefore performed using cytogenetics alone.
  • WGS results were used to assign patients to risk groups using the identical guidelines as above for both AML and MDS patients.
  • risk assignment used CNAs, recurrent SVs, and gene mutations.
  • FLT3 ITD mutation results from PCR were used instead of the WGS result (even though ITD alleles can be detected) because the PCR assay is an FDA-cleared companion diagnostic for the FLT3 targeted therapy midostaurin.
  • the clinically important classifications of normal karyotype and complex karyotype used only CNAs and recurrent SVs and not SVs reported as secondary findings.
  • a normal karyotype was designated if no variants in either category were identified, and a complex karyotype was designated if at least 3 chromosomal abnormalities were identified, including recurrent SVs (not WHO category-defining events) or CNAs greater than 5 Mbp that were identified by copy number analysis and that involved separate chromosome arms. All but 3 of the patients with a complex karyotype could be assigned to this category based on CNAs alone, which indicates that copy number gains and losses are defining features of this phenotype.
  • Cox proportional-hazards regression was used to calculate hazard ratios and test for equal survival between the adverse risk group and either intermediate, favorable, or a combined intermediate/favorable ‘not adverse’ risk group. All log-rank tests performed in the paper were adjusted for multiple comparisons using the method of Benjamini and Hochberg (1995). Cox regression was adjusted for age (binned by decade), which was significantly associated with overall survival in the 71 non-transplanted patients with defined risk stratified by conventional risk groups (HR: 1.46, 95% CI 1.05-2.05) but not WGS-based risk groups (HR: 1.29, 95% CI 0.92-1.81).
  • the log of the white blood cell count was also used as a covariate with ELN risk, but was not significant in any analysis (P>0.05 in all analyses) and therefore was not included in the model.
  • the proportional hazards assumption was found to be tenable for all Cox models.
  • the power calculation used a median survival of 3600 days of survival for the favorable group and 346 for the adverse group, with a minimum follow-up interval of 279 days and a total number of days (accrual+follow-up) of 750 days. This demonstrated 80% power to detect a survival difference between favorable and adverse risk at a sample size of 12 (per group) using an alpha of 0.05. Additional exploratory analyses were performed but not presented, including log-rank tests for differences in survival among all three risk groups (rather than not adverse vs. adverse) and unadjusted Cox regression tests, which yielded similar results to those shown here. Survival statistics were obtained using SAS for Windows, Version 9.4. The survminer package in R was used for visualization.
  • Whole-genome sequencing identified cytogenetically cryptic structural variants in 13 patients, including complex or cryptic chromosomal translocations involving the inv(16)(p13.1q22) fusion gene CBFB-MYH11 in 2 patients, the t(7;21)(p22;q22) fusion gene USP42-RUNX1 in 1 patient, and 10 rearrangements involving KMT2A, all of which were verified with the use of orthogonal methods ( FIG. 6B and FIG.
  • This set of consecutive patients was also evaluated to estimate the diagnostic yield from whole-genome sequencing as compared with testing with cytogenetic analysis and targeted sequencing.
  • This analysis was performed separately in samples obtained from patients with AML and in those obtained from patients with MDS.
  • the comparisons included clinical results from a standard FISH panel along with cytogenetic analysis and targeted sequencing to provide a realistic estimate of the expected yield of whole-genome sequencing.
  • results from conventional cytogenetic analysis and FISH assays in the 68 patients with AML resulted in the diagnosis of acute promyelocytic leukemia with the fusion gene PML-RARA in 5 patients and in the assignment of 27 patients to the adverse-risk group, 10 to the intermediate-risk group, and 19 to the favorable-risk group on the basis of established guidelines; 7 patients had unsuccessful or inconclusive results on cytogenetic analysis and could not be assigned to a risk group.
  • Whole-genome sequencing analysis identified risk-defining chromosomal abnormalities in 4 patients, including KMT2A and RUNX1-RUNXT1 rearrangements in 1 patient each or a complex karyotype in 2 patients; the remaining 23 patients had either a normal karyotype or one or two abnormalities and were assigned to a risk category on the basis of mutations identified by whole-genome sequencing ( FIG. 24 ).

Landscapes

  • Bioinformatics & Cheminformatics (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Evolutionary Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medical Informatics (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Theoretical Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

A computer-implemented method for the identification of clinically relevant structural variants in a subject with AML or MDS from whole genome sequencing data is disclosed that includes providing a whole-genome sequencing dataset, performing a structural variant analysis on the whole-genome sequencing dataset and producing a report that includes clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims priority to U.S. provisional application No. 63/162,665 filed on Mar. 18, 2021, the content of which is incorporated by reference in its entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • Not applicable.
  • MATERIAL INCORPORATED-BY-REFERENCE
  • Not applicable.
  • FIELD OF THE DISCLOSURE
  • The present disclosure generally relates to methods of genomic profiling using whole-genome sequencing.
  • BACKGROUND OF THE DISCLOSURE
  • Chromosome analysis has been used for cancer diagnosis and to guide treatment decisions for over 40 years. The discovery of chromosome-level mutations like the BCR-ABL1 fusion gene in chronic myeloid leukemia (CML) and PML-RARA gene fusion in AML have transformed these once lethal cancers into diseases that can be essentially cured with targeted therapies. Over a thousand chromosome-level mutations have now been identified across numerous cancer types, and although more recent genomic studies of cancer have revealed many additional nucleotide-level cancer-associated mutations, cytogenetic mutations still account for the majority of clinically-relevant genomic changes in cancer. For example, FISH and karyotyping are required for the risk classification system in AML, and cytogenetic testing for chromosomal rearrangements facilitates accurate diagnosis of B-cell lymphomas and guides therapy in non-small cell lung cancer. Moreover, the oncogenes targeted by many of the FDA-approved cancer therapies result from chromosomal rearrangements that are routinely detected using karyotyping or FISH.
  • While effective, conventional cytogenetic methods are imprecise. Karyotyping depends on identifying changes in chromosomes using their unique banding patterns, meaning that small rearrangements can be missed and complex structural mutations may obscure important findings that are clinically actionable. Perhaps the most limiting aspect of conventional cytogenetic testing is that it requires culturing of live cells under stimulating conditions, which makes the method essentially unavailable to the 90% of cancer patients with solid tumors. Certain chromosomal rearrangements can be tested for using FISH, which is routinely performed on formalin-fixed biopsies of solid tumors. However, this approach uses locus-specific DNA probes that can miss rearrangements with atypical breakpoints or identify changes that do not result in the expected mutant gene products and may therefore not result in the anticipated clinical outcome. The most limiting aspect of FISH testing is that it is impractical to test clinical samples for more than a few specific mutations at a time, which can result in testing that is incomplete if sample amounts are insufficient. The need for multiplex testing has led to the development of targeted sequencing panels (e.g., the Foundation of Medicine Comprehensive Cancer Panel, some of which can detect both copy number mutations (e.g., gene deletions and amplifications) and selected gene fusion events. Although these technologies can accurately identify an expanding number of mutations, they require complicated laboratory procedures, often involving both DNA and RNA, and take multiple days to complete. In addition, the assays must predefine the genomic regions that are selected for targeted sequencing and can therefore only identify chromosomal rearrangements that occur at these specific loci. As a result, relatively few tumors are ever tested for the full range of clinically relevant chromosome-level mutations, including those that may respond to approved targeted therapies. To expand precision medicine, new methods are needed that can be applied to more cancer samples and that test for more mutations, including those that are known to predict response to targeted therapies.
  • Genetic profiling is a routine component of the diagnostic workup for an increasing number of cancers and is used to predict clinical outcomes and responses to targeted therapies. Mutations that are clinically actionable for any individual type of cancer typically span a wide range of genomic events, including chromosomal rearrangements, gene amplifications and deletions, and single-nucleotide changes. The diversity of these findings necessitates the use of multiple platforms to obtain the genomic information needed for clinical management. Whole-genome sequencing is an unbiased method of detecting all types of mutations that could potentially be used to replace current testing algorithms. Such sequencing can also be performed on a limited amount of DNA to identify genomic changes that may be cryptic in other types of analyses. These features of whole-genome sequencing suggest that it could improve genomic profiling in patients with cancer.
  • Genomic abnormalities are particularly important for diagnostic classification and risk assessment in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Recurrent chromosomal abnormalities are the basis for the AML genomic classification system of the World Health Organization, and the association of these alterations and certain genetic mutations with clinical outcomes has led to the development of algorithms for genetic risk stratification in patients with AML. Similar studies involving patients with MDS have resulted in the cytogenetic component of the International Prognostic Scoring System-Revised (IPSS-R) in such patients. Although advances in sequencing technology have improved the ability to identify genetic mutations, the detection of chromosomal rearrangements is primarily performed through conventional metaphase cytogenetic analysis (i.e., karyotyping). The latter approach is effective but has several limitations, including the need to obtain viable cells, low sensitivity, and limited resolution.
  • Fluorescence in situ hybridization (FISH) and targeted sequencing assays that use DNA, RNA, or both are also used, but these methods are informative only in the regions selected for analysis and may provide incomplete information regarding identified chromosomal rearrangements. As a result, conventional cytogenetic analysis remains an essential component of the diagnostic workup for patients with AML or MDS.
  • The importance of genetic profiling in such patients and the variety of clinically relevant mutation types suggest that whole-genome sequencing could be used in place of standard testing approaches. Although the high cost of sequencing and complex, time-consuming analysis methods have historically restricted such sequencing to research studies, recent advances have made this analysis simpler to perform, faster, and less expensive.
  • Other objects and features will be in part apparent and in part pointed out hereinafter.
  • SUMMARY OF THE DISCLOSURE
  • In one aspect, a computer-implemented method for the identification of clinically relevant structural variants in a subject with AML or MDS from whole genome sequencing data is disclosed that includes providing a whole-genome sequencing dataset, the whole-genome sequencing dataset comprising a plurality of alignments of tumor DNA sequence fragments to a reference human genome to a computing device; performing, using the computing device, a structural variant analysis on the whole-genome sequencing dataset, the structural variant analysis including copy-number alteration (CNA) identification, structural variant (SV) identification, and gene-level variant identification to identify clinically relevant structural variants indicative of AML or MDS within the whole-genome sequencing dataset; and producing, using the computing device, a report comprising the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis. In some aspects, copy-number alteration (CNA) identification further comprises transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of read counts over 500,000 bp nonoverlapping windows across the genome; transforming, using the computing device, the plurality of read counts into a plurality of CNAs; and filtering, using the computing device, plurality of CNAs to retain only CNAs greater than 5 Mbp. In some aspects, SV identification further comprises: transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of SV calls; filtering, using the computing device, the plurality of SVs to retain only SV calls greater than 100 kbp in length; and filtering, using the computing device, the SV calls greater than 100 kbp in length to identify translocations, deletions, duplications, and inversions that overlap a predefined list of recurrent and/or risk-defining SVs associated with AML or MDS. In some aspects, gene-level variant identification further comprises identifying, using the computing device, the alignments of the whole-genome sequencing dataset within about 85 kbp targeting 40 predetermined genes and gene hotspots that are recurrently mutated in AML or MDS. In some aspects, the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis are indicative of a clinical outcome of the subject. In some aspects, providing the whole-genome sequencing dataset whole genome sequencing data further comprising performing whole-genome sequencing on a biological sample comprising tumor DNA from the subject with about 60× genome coverage.
  • DESCRIPTION OF THE DRAWINGS
  • The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
  • Those of skill in the art will understand that the drawings, described below, are for illustrative purposes only. The drawings are not intended to limit the scope of the present teachings in any way.
  • FIG. 1 is a block diagram schematically illustrating a system in accordance with one aspect of the disclosure.
  • FIG. 2 is a block diagram schematically illustrating a computing device in accordance with one aspect of the disclosure.
  • FIG. 3 is a block diagram schematically illustrating a remote or user computing device in accordance with one aspect of the disclosure.
  • FIG. 4 is a block diagram schematically illustrating a server system in accordance with one aspect of the disclosure.
  • FIG. 5 is a schematic illustration of the workflow and approximate processing time for each step of the rapid WGS method and analysis of samples obtained from the study patients. (An example of the reports that were generated by this process are provided in FIG. 9).
  • FIG. 6A is a comparison of WGS with Conventional Cytogenetic Analysis and Targeted Gene Sequencing. It shows the sensitivity of WGS for the detection of recurrent structural variants (SVs) and copy-number alterations (CNAs) as compared with conventional cytogenetic analysis and for the detection of single-nucleotide variants (SNVs) and insertion-deletions (INDELs) as compared with high-coverage targeted gene sequencing. Error bars denote 95% confidence intervals.
  • FIG. 6B shows the identification and confirmation by WGS of 13 new recurrent SVs that were not detected by conventional cytogenetic analysis, as supported by orthogonal methods, including fluorescence in situ hybridization (FISH), polymerase chain reaction (PCR) with sequencing of SV breakpoints, or detection of fusion transcripts in RNA-sequence (RNA-seq) data.
  • FIG. 6C shows the identification of 21 new CNAs in 14 patients; 12 of these alterations were confirmed by chromosomal microarray (CMA), FISH, or sequence-defined breakpoints. An additional 9 CNAs were identified by WGS only and could not be confirmed by CMA (in 6 patients) or confirmation was not attempted because of the size or abundance of the CNA event (in 3 patients). CNAs were also identified in 13 patients with ambiguous or inconclusive results on cytogenetic analysis. Additional details regarding these comparisons are provided in Tables S4 and S5 and FIGS. 18A, 18B, 18C, and 18D.
  • FIG. 7A describes is a bar graph summarizing the time it took to perform WGS-Based Genomic Profiling on samples obtained from 117 consecutive patients with AML or MDS by means of WGS, as indicated by the dashed horizontal black line. The height of each bar shows the total time in days for processing, starting from construction of the sequencing library and ending with completion of the automated final report for an individual patient sample. The duration of each individual step (as obtained from time stamps recorded in the information management system of the clinical laboratory) is indicated by the shaded bar segments and includes the duration of library generation and quality assessment, sequencing, and analysis and reporting. These times reflect the processing time plus waiting time before the next step. Longer turnaround times occurred because of delays between steps, rather than longer processing times. The dashed horizontal red lines show the recommended maximum turnaround time for FISH testing and conventional cytogenetic analysis, according to published recommendations, although shorter turnaround times occur in many laboratories.
  • FIG. 7B describes the Diagnostic Yield of WGS-based Genomic Profiling in 117 Consecutive Patients. It shows the yield of new WGS findings in samples obtained from 68 unselected, consecutive patients with AML. FIG. 7B shows the cumulative number of patients with new genomic findings that were identified by WGS, as compared with conventional cytogenetic analysis or FISH, performed at the time of diagnosis, along with the cumulative number of patients with new events that changed the category of genetic risk group on the basis of established European Leukemia Network (ELN) guidelines. FISH testing included assays for PML-RARA, CBFB-MYH11, RUNX1-RUNX1T1, del(5q), and chromosome 7 deletion, according to recommendations; all testing was performed in samples obtained from 60 of 68 patients (88%); subgroups of these assays were performed for the remaining patients. The results of ELN assignments to a genetic risk group by WGS, conventional cytogenetic analysis with FISH, and cytogenetic analysis alone are shown in the top panel. The red asterisk indicates that the patient's risk group was reclassified according to the WGS results, and the red arrow indicates that the risk-group assignment was based on FISH results alone.
  • FIG. 7B shows the genomic events that were detected by WGS and are labeled as concordant with cytogenetic analysis, with FISH, or with target sequencing (in black), new findings made by WGS (in blue), and new findings that resulted in a change in the ELN genetic risk group (in red). The status regarding internal tandem duplication in FLT3 (FLT3-ITD) and the allele ratio as determined by PCR were used for both conventional and WGS-based risk stratifications.
  • FIG. 8A is a risk assessment by WGS in Patients with AML, according to Existing Genetic Risk Groups. It shows overall survival for 71 patients with AML who were treated with chemotherapy alone after remission, as stratified into established ELN genetic risk groups on the basis of a combination of conventional cytogenetic analysis, FISH, and targeted gene sequencing.
  • FIG. 8B shows the same cohort as in FIG. 8A with risk stratification according to WGS results. The ratio of the mutated FLT3-ITD allele to the wild-type allele, as determined by PCR, was used for both the conventional and WGS classifications; the presence or absence of the mutation was used when allele ratios were not available.
  • FIG. 8C shows the clinical outcomes for 27 patients for whom genetic risk could not be determined because of inconclusive, unsuccessful, or unknown results on cytogenetic analysis. The median survival in this cohort was 11.2 months (95% confidence interval [CI], 5.6 to 38.8).
  • FIG. 8D shows the stratification of the cohort in FIG. 8C into established genetic risk groups with the use of WGS results, which predicted shorter overall survival for patients at adverse risk than for those at intermediate or favorable risk (not adverse) (age-adjusted hazard ratio for death for intermediate or favorable risk versus adverse risk, 0.29; 95% CI, 0.09 to 0.94). All P values were calculated with the use of a log-rank test for equal survival among the groups and adjusted for multiple comparisons.
  • FIG. 9 is an exemplary cover page of a graphical ChromoSeq WGS report highlighting the clinically significant findings in the genome sequence of an AML patient.
  • FIG. 10 is a histogram distribution of genome coverage of the genome-wide coverage depth in unique reads for 235 WGS cases.
  • FIG. 11 summarizes the variants detected per patient from a number distribution of SVs, CNAs, and gene mutations detected in 235 patients.
  • FIG. 12 is series of images confirming the SVs detected using the disclosed WGSA method. The images show FISH results from metaphase and interphase FISH analysis utilizing a dual color, break apart probe targeting KMT2A (manufactured by Vysis). A normal signal hybridization pattern is 2Y. Rearrangement of a KMT2A locus generates a separation of the green signal, which encompasses the 5′ segment of KMT2A and surrounding region, and the red signal, which encompasses the 3′ segment of KMT2A and surrounding region.
  • FIG. 13 is a series of images showing selected CNAs obtained by FISH. See also Table S5.
  • FIG. 14A is a first normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 14B is a second normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 14C is a third normalized coverage plot of WGS results for CNAs that were detected by WGS but could not be confirmed because of their small size, low abundance, lack of FISH probes, or lack of material.
  • FIG. 15A is a first normalized coverage plot of WGS results for CNAs that were detected by WGS.
  • FIG. 15B is a second normalized coverage plot of WGS results for CNAs that were detected by WGS.
  • FIG. 16 shows the sensitivity of WGS for mutations in AML risk-defining genes. Gold standard mutations NPM1 (NPM1c only), CEBPA, FLT3 (non-ITD mutations), RUNX1, and TP53 were obtained from targeted sequencing (>500× coverage) using a clinical assay (N=103 patients). FLT3-ITD mutations were obtained from clinical testing via PCR and capillary electrophoresis (N=104 patients). Numbers above each bar indicate the sensitivity (TP/(TP+FN)×100) and the number of true positives and total positives by the gold standard assay.
  • FIG. 17 compares FLT3-ITD detection by WGS vs. PCR. Results for 35 patients with FLT3-ITD mutations detected by either PCR and capillary electrophoresis or WGS. Rows show results for each patient with a positive ITD assay, including WGS, a clinical targeted sequencing assay, and PCR. The ITD allele sizes and allele ratios are indicated for each assay. Allele sizes and ratios were available for all positive results. Data from PCR include an in-house laboratory developed test (LDT) and the commercial FDA companion diagnostic assay, and therefore the allele size and ratios were not always reported. Note that ITD alleles were detected by WGS in two patients for whom the in-house LDT assay was negative (in 2002). ITD alleles were also detected in these patients in the AML TCGA study).
  • FIG. 18A shows the VAFs from deep targeted sequencing for 348 variants detected in 102 patients using a clinical targeted sequencing assay. Left panel shows the VAF from targeted sequencing for variants detected (N=300, in blue) and missed (N=48, in orange) by WGS.
  • FIG. 18B shows the distribution of VAFs for the detected and missed variants of FIG. 18A.
  • FIG. 18C shows the abundance and coverage depth of gene mutations in WGS data for 348 variants detected in 102 patients using a clinical targeted sequencing assay. Left panel shows WGS coverage for variants detected (N=300, in blue) and missed (N=48, in orange) by WGS. Right panels show the distribution of WGS coverage for detected and missed variants.
  • FIG. 18D shows the distribution of VAFs for the detected and missed variants of FIG. 18C.
  • FIG. 19A is a coverage metric for false negative gene mutations. It shows WGS coverage (indicated by the height of the bar) and variant supporting reads (height of the blue bar) for 48 variants that were detected by clinical targeted sequencing but missed by WGS. Most of the variants (41/48) were present in the WGS data, but at very low frequency. Detection of gene variants required >3 variant-supporting reads and >6× total coverage. Variants highlighted by the asterisk were subsequently detected upon top-up sequencing of 4 cases.
  • FIG. 19B is a coverage metric for false negative gene mutations. It demonstrates the theoretical binomial sampling probability for detecting variants at VAFs ranging from 2% to 20% and coverage levels from 0 to 100×. Note that at 50× coverage (the mean obtained for this study), there is a 45% probability of sampling a variant >3 times if the true abundance is 5%. This is consistent with previous work showing that ‘standard coverage’ WGS is inadequate for robust detection of low frequency variants, but this can be improved by increasing coverage depth (see also FIG. 20).
  • FIG. 20 shows additional variants detected after top-up sequencing. WGS missed 11 gene variants from patients 312088, 681540, 416413, and 262878 that were either present at low abundance in targeted sequencing data (<20% VAF, N=7) or occurred at position with low coverage in the WGS data (<25×, N=4). Additional sequencing of these samples increased the coverage for these samples from a mean of 35× (range: 17×-58×) to 83× (range: 59×-121×), and resulted in the detection of 9 of the 11 missed variants. The remaining 2 variants were present at low frequency in the WGS data but were not detected by the variant analysis pipeline.
  • FIG. 21A is the diagnostic yield from prospective sequencing of 42 consecutive MDS patients with WGS. FIG. 21A is organized as in FIG. 7B. Consecutive MDS patients were sequenced from April 2019 to February 2020 to estimate the diagnostic yield of WGS compared to standard testing. Top panel shows the cumulative number of patients with new findings (in blue) and the cumulative number of patients with findings that changed the IPSS-R cytogenetic risk score. IPSS-R from cytogenetics and sequencing are shown below.
  • FIG. 21B shows chromosomal abnormalities obtained from WGS and indicates new findings (in blue) and findings that changed the IPSS-R category (red) in top panel. Bottom panel shows mutations in MDS-associated genes referenced in NCCN guidelines, with concordance between WGS and targeted sequencing indicated by the color (concordant in black, WGS only in orange, and targeted sequencing only in green). Note that the yield in risk-defining events in this cohort is due entirely to cases where cytogenetics was unsuccessful or inconclusive, resulting in an undetermined IPSS-R risk category.
  • FIG. 22A represents AML patients with reclassified risk groups by WGS. AML patients included in the outcome analysis were with defined cytogenetic risk that were reclassified by WGS. It shows the ELN risk groups by cytogenetics combined with FISH for PML-RARA, RUNX1-RUNX1T1, CBFB-MYH11, del(5q), and del(7q) (bottom), and gene mutation testing either via targeted sequencing or PCR, and WGS and FLT3-ITD PCR only (top).
  • FIG. 22B show WGS results (with the risk-defining event highlighted in red), and results from cytogenetics, FISH, and gene mutation analysis for AML patients with reclassified risk groups by WGS. FLT3-ITD mutation status by PCR was used for both WGS and conventional risk group assignments. Cells outlined in red indicate that testing was either not performed or failed. Also shown is the clinical status of each patient, including whether they expired or relapsed, and the follow-up time in months from diagnosis. WGS identified new adverse risk findings in 5 patients, while 3 patients had differences in gene mutations in ASXL1 and NPM1 and either due to lack of testing at diagnosis (N=2) or a missed low abundance NPM1c mutation by WGS (N=1).
  • FIG. 23A is a survival analysis of 101 AML patients with defined risk. It shows Kaplan Meier survival curves using death as the endpoint for 101 AML patients treated with either post-remission chemotherapy alone (N=71) or allogeneic stem cell transplant (N=30) stratified by ELN risk groups from cytogenetics, targeted sequencing, and FLT3 ITD mutation testing. Log-rank test for equal survival across the groups, adjusted P=0.43. Age adjusted Cox regression for death in not adverse vs. adverse cytogenetic risk groups: 1.06, 95% CI 0.45 to 2.50.
  • FIG. 23B shows Kaplan Meier survival curves using death as the endpoint for 101 AML patients treated with either post-remission chemotherapy alone (N=71) or allogeneic stem cell transplant (N=30) stratified by ELN risk groups from WGS and FLT3 ITD mutation testing. Log-rank test for equal survival across the groups, adjusted P=0.09. Age-adjusted Cox regression for death in not adverse and adverse WGS-based risk groups: 0.59, 95% CI 0.26 to 1.36.
  • FIG. 24A are WGS results for AML patients with inconclusive cytogenetics, or patients with unsuccessful (N=6), inconclusive (N=13), or unknown (N=8) cytogenetic and FISH studies that precluded definitive genomic risk assignment. WGS-based ELN risk group is shown in the top panel.
  • FIG. 24B are results from WGS, cytogenetics, FISH, and gene mutation testing. FLT3-ITD mutation status was determined by PCR and was used for risk stratification according to ELN criteria using an allele ratio cutoff of 0.5, or presence/absence if an allele ratio was not available. Bottom panel shows clinical status and follow-up time in months. WGS identified risk-defining chromosomal abnormalities in four patients, including KMT2A and RUNX1-RUNX1T1 rearrangements (N=1 each) and a complex karyotype (N=2). The remaining 23 patients were assigned to ELN risk groups based on gene mutations.
  • FIG. 25A is a survival analysis of AML patients with inconclusive cytogenetics, showing a Kaplan Meier survival curve for 27 AML patients with unsuccessful for inconclusive cytogenetics stratified by gene mutations only. Patients were considered intermediate risk unless favorable risk or adverse risk gene mutations were identified. Log-rank test for equal survival across the groups, adjusted P=0.09. Age-adjusted Cox regression hazard ratio for death in not adverse vs. adverse risk, 0.4; 95% CI, 0.14 to 1.1.
  • FIG. 25B is a Kaplan Meier survival curve using death as the endpoint for the above cohort of 38 patients stratified by ELN risk groups from WGS and FLT3 ITD mutation testing. Median survival for this expanded cohort was 22.3 months, 95% CI, 6.8 to 46.1. Log-rank test for equal survival across the groups, adjusted P=0.02. Age-adjusted Cox regression hazard ratio for death in not adverse vs. adverse risk, 0.28; 95% CI, 0.11 to 0.71.
  • There are shown in the drawings arrangements that are presently discussed, it being understood, however, that the present embodiments are not limited to the precise arrangements and are instrumentalities shown. While multiple embodiments are disclosed, still other embodiments of the present disclosure will become apparent to those skilled in the art from the following detailed description, which shows and describes illustrative aspects of the disclosure. As will be realized, the invention is capable of modifications in various aspects, all without departing from the spirit and scope of the present disclosure. Accordingly, the drawings and detailed description are to be regarded as illustrative in nature and not restrictive.
  • DETAILED DESCRIPTION
  • In various aspects, a rapid method based on whole-genome sequencing that recapitulates and improves upon conventional cytogenetics is disclosed. Whole-genome sequencing (WGS) can detect clinically relevant chromosomal rearrangements with unparalleled accuracy and would vastly improve karyotyping of tumors by making it possible to test nearly any tumor type (including formalin-fixed specimens) for virtually all clinically-relevant chromosomal rearrangements simultaneously. The methods disclosed herein advance 50-year-old cytogenetic methods to the modern era by greatly expanding the range of mutations that can be detected and tumor types that can be analyzed with the ultimate goal of improving clinical decisions and patient outcomes. The disclosed method includes both software and wet laboratory workflows.
  • Genetic profiling is a routine component of the diagnostic workup for an increasing number of cancers and is used to predict clinical outcomes and responses to targeted therapies. Mutations that are clinically actionable for any individual type of cancer typically span a wide range of genomic events, including chromosomal rearrangements, gene amplifications and deletions, and single-nucleotide changes. The diversity of these findings necessitates the use of multiple platforms to obtain the genetic information needed for clinical management. Whole-genome sequencing is an unbiased method of detecting all types of mutations and could potentially be used to replace current testing algorithms. Such sequencing can also be performed on a limited amount of DNA and can identify genomic changes that may be cryptic in other types of analyses. These features of whole-genome sequencing make the methods disclosed herein suitable for genomic profiling in patients with cancer.
  • At least several features of WGS make it particularly well-suited for use in clinical testing. WGS can be performed using minimal amounts of DNA (as little as 50 ng) and from any specimen type, including formalin-fixed solid tumor tissue obtained from routine surgical biopsies, and so can be applied to nearly any tumor type and is not limited to only those tumor types with live cells. WGS procedures also do not involve complicated and time-consuming laboratory steps that are required for other sequencing methods, including complex library preparation procedures, hybridization-capture enrichment, or intact RNA, making it one of the most rapid next-generation sequencing assays and among the simplest to implement in a clinical laboratory. Finally, because WGS produces genome-wide, base-pair resolution genomic data, it can be analyzed for chromosomal rearrangements and gene-level mutations simultaneously, thereby providing a comprehensive profile of all clinically relevant mutations regardless of mutation type.
  • In various aspects, the disclosed method includes providing or obtaining a biological sample comprising tumor DNA. Any suitable biological sample may be used in the disclosed method without limitation including, but not limited to, peripheral blood, bone marrow aspirate, solid tumor biopsy samples, and any other suitable biological sample. The sample size may be at least about 200 μL for samples such as peripheral blood and bone marrow aspirate.
  • To reduce time, complexity, and cost, the disclosed method is implemented without a normal tissue comparator because the method is configured to identify clearly pathogenic somatic events that generally do not require a germline control. In some aspects, the biological sample is provided as previously-obtained WGS data. In other aspects, method includes extracting the tumor DNA from the biological sample using any suitable method without limitation including, but not limited to, the QIAamp DNA mini kit (Qiagen, Hilden, Germany) as detailed in the package insert, followed by quantification with the Qubit 1.0 fluorometer High Sensitivity dsDNA assay (ThermoFisher, Waltham, Mass.) as described in the Example below.
  • In various aspects, the method further includes performing library preparation using the tumor DNA extracted from the biological sample. In some aspects, the amount of tumor DNA used for library preparation ranges from about 35 ng to about 500 ng or more. Any suitable method of library preparation may be used without limitation including, but not limited to the Nextera Flex library preparation kit (cat #20015804, Illumina, Inc, San Diego, Calif.) as described in the Examples below. In some aspects, the quantified final libraries are diluted to 1 nM for equimolar pooling prior to sequencing
  • In various aspects, the method further includes subjecting the library to whole-genome sequencing using any suitable systems and associated methods without limitation. In one exemplary aspect, WGS is performed using a NovaSeq 6000 sequencing instrument (Illumina) configured to obtain about 60× genome coverage per sample. In various other aspects, the WGS may be configured to obtain a genome coverage ranging from about 10× per sample to about 100× per sample. In other aspects, the WGS may be configured to obtain a genome coverage of about 10×, 20×, 30×, 40×, 50×, 60×, 70×, 80×, 90×, 100×, or more per sample.
  • In various aspects, the method further includes aligning the WGS data to a human reference genome including, but not limited to the GRCh38 human reference genome. Any suitable alignment method may be used without limitation including local alignment software such as DRAGEN (version 3.5.7) hardware-accelerated sequence processing software suite or cloud-based alignment software such as the DRAGEN Germline (alignment only) BaseSpace App. In various aspects, the alignments are provided for variant analysis in any suitable format including, but not limited to CRAM format.
  • In various aspects, the disclosed method makes use of a streamlined approach for rapid whole-genome sequencing and analysis that detects clinically relevant cytogenetic abnormalities in a range of tumor types. In various aspects, the ChromoSeq WGS assay is a high-performance tumor-only WGS analysis pipeline that generates a digital karyotype and detects known chromosomal rearrangements and gene-level mutations based on analysis of raw WGS sequence data produced from a single biological sample.
  • In various aspects, the method further includes performing variant analysis on the alignments using the ChromoSeq WGS assay. Variant analysis includes CNA identification, SV identification, and gene-level variant identification. In some aspects, each portion of the variant analysis is limited to targeted analysis and filtering to streamline the process while yielding variants that are be clinically relevant. In various aspects, each portion of the variant analysis is performed using the same alignments from the single biological sample.
  • For CNA identification, the alignments are transformed into read counts in 500,000 bp nonoverlapping windows across the genome and the read counts are transformed into CNAs using any suitable method including, but not limited to, the purity and subclone-aware Hidden Markov Model, ichorCNA as described in the examples below. In some aspects, the reported CNAs are filtered to remove CNAs <5 Mbp; cytogenetically evident CNAs are typically greater than plurality of CNAs, a size potentially detectable by karyotype analysis.
  • For SV identification, a break-end caller Manta is used to detect SV calls of at least 100 kbp in length to reduce the number of calls with unknown clinical significance. The detected SVs are then filtered to identify translocations, deletions, duplications, and inversions that overlap a curated list of 612 recurrent and/or risk-defining SVs obtained from published sources, including the WHO and the Atlas of Genetics and Cytogenetics in Oncology. A list of the recurrent and/or risk-defining SVs is provided as Table S2 in the examples below. Genomic events where both ends overlap one of these recurrent SVs are reported as ‘top-level’ findings in ChromoSeq without additional filtering. The remaining SVs are subsequently filtered to remove patient-specific events and/or identify cytogenetically cryptic rearrangements involving genes relevant for AML or MDS. In some aspects, the filtering criteria may include retaining those SVs based on: 1) at least 2 ‘paired and 2 ‘split’ reads supporting the break-ends, 2) absence of an overlapping call from a large set of SVs identified from >17,795 human genomes, 3) coverage depth of deletion or duplication call must be <0.8 or >1.3 compared to the background, respectively, and 4) a defined breakpoint must be identified and the spanning contig generated by Manta must map back to the reported breakpoints.
  • Gene-level variants are identified using the same alignments from the biological sample as used for CNA and SV analysis. In some aspects, gene mutations are identified within about 85 kbp targeting 40 genes and gene hotspots that are recurrently mutated in AML or MDS. In other aspects, an indel caller is run on exons 13-15 of FLT3 to identify FLT3 ITD alleles.
  • In various aspects, the method includes combining the annotated CNA, SV, and gene mutation calls obtained as described above are combined with coverage QC information to generate a final text report as well as data files (CRAM, and VCF) and graphical coverage plots from ichorCNA. In some aspects, a graphical report is generated, as shown in FIG. 9.
  • In various aspects, the disclosed method detects clinically significant structural variants (copy number alterations and translocations) from WGS data, and provides the ability to replicate and outperform conventional cytogenetic testing. The ChromoSeq WGS automated workflow sifts down up to 10,000 or more potential CAN and SV calls to 1 or 2 real events without sequencing paired normal tissue, greatly streamlining and simplifying WGS for use as a clinical assay.
  • In various aspects, the disclosed methods are streamlined at each phase to provide for clinically timely results. In some aspects, scalable methods of sample preparation that can be performed by a single technician in less than 8 hours with commercially available reagents are used. In other aspects, the samples are subjected to high-throughput sequencing followed by automated tumor-only variant analysis to detect mutations in selected genes, copy-number alterations of more than 5 Mbp, and recurrent structural variants. In additional aspects, the method includes automatically generating the findings of the analysis in a concise clinical report.
  • In addition to improving on existing clinical methods such as conventional cytogenetics, the methods disclosed herein obviate at least a portion of the challenges associated with the use of WGS for clinical testing, in particular the high cost of sequencing and the complex analysis of results that are typically too complicated and time consuming for clinical laboratories. In some aspects, the disclosed methods include a simplified, tumor-only WGS analysis strategy that focuses on detecting known, clinically-relevant chromosome-level mutations that are routinely tested for by clinical cytogenetic laboratories. By limiting WGS analysis to mutations with established clinical relevance, the disclosed method greatly reduces the time, cost, and complexity of WGS, while also expanding the number of mutations that can be queried in each sample. Previous research of tumors using WGS have been focused on comprehensive discovery of new mutations, rather than efficient and comprehensive detection of known mutations.
  • In other aspects, the disclosed method makes use of recently-developed sequencing systems including, but not limited to, the Illumina NovaSeq 6000 instrument to perform WGS of tumor samples. Such systems are capable of generating high coverage WGS data in a matter of days at a cost that is comparable to standard karyotyping and cytogenetic analysis that is typically used for clinical testing.
  • In various aspects, a streamlined approach to whole-genome sequencing (ChromoSeq) is disclosed. The disclosed method is designed to provide comprehensive genomic profiling of clinically relevant mutations in samples obtained from patients with AML or MDS, while minimizing the turnaround time and technical complexity. An overview of the disclosed method is provided in FIG. 5. As illustrated in FIG. 5, scalable methods of sample preparation that can be performed by a single technician in less than 8 hours with commercially available reagents were used, followed by standard high-throughput sequencing. Automated tumor-only variant analysis detected mutations in selected genes, copy-number alterations of more than 5 Mbp, and recurrent structural variants (Tables S1 and S2). The findings of the WGS analysis is summarized in a concise clinical report to a practitioner (FIG. 9).
  • As described in the Examples below, that whole-genome sequencing provided rapid and accurate genomic profiling in patients with AML or MDS. WGS sequencing also provided a greater diagnostic yield than conventional cytogenetic analysis and more efficient risk stratification on the basis of standard risk categories.
  • Genomic abnormalities are particularly important for diagnostic classification and risk assessment in patients with acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). Recurrent chromosomal abnormalities are the basis for the AML genomic classification system of the World Health Organization, and the association of these alterations and certain genetic mutations with clinical outcomes has led to the development of algorithms for genetic risk stratification in patients with AML. Similar studies involving patients with MDS have resulted in the cytogenetic component of the International Prognostic Scoring System-Revised (IPSS-R) in such patients. Although advances in sequencing technology have improved the ability to identify genetic mutations, the detection of chromosomal rearrangements is primarily performed through conventional metaphase cytogenetic analysis (i.e., karyotyping). The latter approach is effective but has several limitations, including the need to obtain viable cells, low sensitivity, and limited resolution. Fluorescence in situ hybridization (FISH) and targeted sequencing assays that use DNA, RNA, or both are also used, but these methods are informative only in the regions selected for analysis and may provide incomplete information regarding identified chromosomal rearrangements. As a result, conventional cytogenetic analysis remains an essential component of the diagnostic workup for patients with AML or MDS.
  • In various aspects, the method includes the use of whole-genome sequencing in place of standard testing for genetic profiling in AML and MDS patients. Although the high cost of sequencing and complex, time-consuming analysis methods have historically restricted such sequencing to research studies, recent advances have made this analysis simpler to perform, faster, and less expensive. As described in the Examples below, the method includes a streamlined approach to whole-genome sequencing for genomic profiling of patients with AML or MDS.
  • As described in the examples below, the clinical utility of whole-genome sequencing for the genomic evaluation of patients with AML or MDS was demonstrated. Results from 263 patients showed that such sequencing was equivalent to or better than conventional testing, both in analytical performance and clinical applicability. Whole-genome sequencing detected 100% of the clinically significant abnormalities that had been identified by the existing clinical methods, cytogenetic analysis and clinical FISH assays. In addition, whole-genome sequencing provided new genetic information in 25% of patients, more than half of whom would have been assigned to a different genetic risk category with results from conventional testing.
  • In some aspects, the diagnostic yield of whole-genome sequencing will depend on laboratory-specific karyotyping practices and the use of FISH or other ancillary testing; and some rapid diagnostic assays may still be used for urgent treatment decisions (e.g., FISH or quantitative PCR for PML-RARA rearrangements and PCR for FLT3-ITD mutations). However, the Examples below demonstrate that whole-genome sequencing provides definitive results for clinically relevant genomic events with the use of a single test.
  • Prospective real-time sequencing of samples obtained from consecutive patients, described below, showed that whole-genome sequencing yields complete genomic information in a clinically relevant timeframe. This speed resulted from faster laboratory methods and automated data analysis that focused on clinically relevant mutations, which allowed the generation of reports in as little as 3 days. In some aspects, WGS results are suitable for use in risk predictions with existing, clinically validated risk-stratification systems. The disclosed method adds prognostic value by expanding risk stratification to more patients, especially for those with inconclusive results on cytogenetic analysis, where whole-genome sequencing could have an immediate effect on treatment decisions.
  • Implementing whole-genome sequencing for clinical testing can provide a unified, stable, and extensible platform that minimizes laboratory-specific bias and that can be standardized throughout the world. Although the disclosed method is described herein for use in diagnosing myeloid cancers, many of the advantages of whole-genome sequencing directly apply to patients with other cancers. Whole-genome sequencing can be performed on DNA from tissue biopsy samples of solid tumors, which are often insufficient for standard molecular assays and difficult to culture for cytogenetic studies. The benefits of WGS may be even greater for these cancer types, in which whole-genome sequencing could be used to rapidly survey the entire genome for an expanding number of key mutations and structural alterations with only a small amount of DNA. Such an approach would simplify genomic testing for these patients and probably increase the yield of clinically relevant findings, which improve the precision of approaches for treating many patients with cancer.
  • In various aspects, at least a portion of the disclosed whole-genome sequencing methods may be implemented using various computing systems and devices as described below.
  • FIG. 1 depicts a simplified block diagram of a computing device for implementing the methods described herein. As illustrated in FIG. 1, the computing device 300 may be configured to implement at least a portion of the tasks associated with the disclosed method using a whole-genome sequencing system 310 including, but not limited to: operating the sequencing system 310 to obtain whole-genome sequencing (WGS) data, analyzing the WGS data to identify mutations, copy-number alterations, structural variants, and generating a clinical report of findings. The computer system 300 may include a computing device 302. In one aspect, the computing device 302 is part of a server system 304, which also includes a database server 306. The computing device 302 is in communication with a database 308 through the database server 306. The computing device 302 is communicably coupled to the sequencing system 310 and a user-computing device 330 through a network 350. The network 350 may be any network that allows local area or wide area communication between the devices. For example, the network 350 may allow communicative coupling to the Internet through at least one of many interfaces including, but not limited to, at least one of a network, such as the Internet, a local area network (LAN), a wide area network (WAN), an integrated services digital network (ISDN), a dial-up-connection, a digital subscriber line (DSL), a cellular phone connection, and a cable modem. The user-computing device 330 may be any device capable of accessing the Internet including, but not limited to, a desktop computer, a laptop computer, a personal digital assistant (PDA), a cellular phone, a smartphone, a tablet, a phablet, wearable electronics, smartwatch, or other web-based connectable equipment or mobile devices.
  • In other aspects, the computing device 302 is configured to perform a plurality of tasks associated with the disclosed whole-genome sequencing method. FIG. 2 depicts a component configuration 400 of computing device 402, which includes database 410 along with other related computing components. In some aspects, computing device 402 is similar to computing device 302 (shown in FIG. 1). A user 404 may access components of computing device 402. In some aspects, database 410 is similar to database 308 (shown in FIG. 1).
  • In one aspect, database 410 includes sequencing data 418 and algorithm data 420. Non-limiting examples of suitable sequencing data 418 include any data associated with the whole genome sequencing and alignment. Non-limiting examples of suitable algorithm data 420 include any values of parameters defining the analysis of the whole genome sequencing data, such as any of the parameters defining the WGS library, variant analysis, copy-number alteration identification, and structural variant analysis. Other non-limiting examples of suitable algorithm data 420 include any parameters defining the formatting of a clinical report of results.
  • Computing device 402 also includes a number of components that perform specific tasks. In the exemplary aspect, computing device 402 includes a data storage device 430, segmentation component 440, analysis component 450, and communication component 460. Data storage device 430 is configured to store data received or generated by computing device 402, such as any of the data stored in database 410 or any outputs of processes implemented by any component of computing device 402. Sequencing component 440 is configured to operate or produce signals configured to operate, a sequencing system to obtain and align whole-genome sequencing data. Analysis component 450 is configured to analyze the WGS data and generate clinical reports as described herein.
  • Communication component 460 is configured to enable communications between computing device 402 and other devices (e.g. user computing device 330 and sequencing system 310, shown in FIG. 1) over a network, such as network 350 (shown in FIG. 1), or a plurality of network connections using predefined network protocols such as TCP/IP (Transmission Control Protocol/Internet Protocol).
  • FIG. 3 depicts a configuration of a remote or user-computing device 502, such as user computing device 330 (shown in FIG. 1). Computing device 502 may include a processor 505 for executing instructions. In some aspects, executable instructions may be stored in a memory area 510. Processor 505 may include one or more processing units (e.g., in a multi-core configuration). Memory area 510 may be any device allowing information such as executable instructions and/or other data to be stored and retrieved. Memory area 510 may include one or more computer-readable media.
  • Computing device 502 may also include at least one media output component 515 for presenting information to a user 501. Media output component 515 may be any component capable of conveying information to user 501. In some aspects, media output component 515 may include an output adapter, such as a video adapter and/or an audio adapter. An output adapter may be operatively coupled to processor 505 and operatively coupleable to an output device such as a display device (e.g., a liquid crystal display (LCD), organic light emitting diode (OLED) display, cathode ray tube (CRT), or “electronic ink” display) or an audio output device (e.g., a speaker or headphones). In some aspects, media output component 515 may be configured to present an interactive user interface (e.g., a web browser or client application) to user 501.
  • In some aspects, computing device 502 may include an input device 520 for receiving input from user 501. Input device 520 may include, for example, a keyboard, a pointing device, a mouse, a stylus, a touch-sensitive panel (e.g., a touchpad or a touch screen), a camera, a gyroscope, an accelerometer, a position detector, and/or an audio input device. A single component such as a touch screen may function as both an output device of media output component 515 and input device 520.
  • Computing device 502 may also include a communication interface 525, which may be communicatively coupleable to a remote device. Communication interface 525 may include, for example, a wired or wireless network adapter or a wireless data transceiver for use with a mobile phone network (e.g., Global System for Mobile communications (GSM), 3G, 4G or Bluetooth) or other mobile data network (e.g., Worldwide Interoperability for Microwave Access (WIMAX)).
  • Stored in memory area 510 are, for example, computer-readable instructions for providing a user interface to user 501 via media output component 515 and, optionally, receiving and processing input from input device 520. A user interface may include, among other possibilities, a web browser and client application. Web browsers enable users 501 to display and interact with media and other information typically embedded on a web page or a website from a web server. A client application allows users 501 to interact with a server application associated with, for example, a vendor or business.
  • FIG. 4 illustrates an example configuration of a server system 602. Server system 602 may include, but is not limited to, database server 306 and computing device 302 (both shown in FIG. 1). In some aspects, server system 602 is similar to server system 304 (shown in FIG. 1). Server system 602 may include a processor 605 for executing instructions. Instructions may be stored in a memory area 625, for example. Processor 605 may include one or more processing units (e.g., in a multi-core configuration).
  • Processor 605 may be operatively coupled to a communication interface 615 such that server system 602 may be capable of communicating with a remote device such as user computing device 330 (shown in FIG. 1) or another server system 602. For example, communication interface 615 may receive requests from user computing device 330 via a network 350 (shown in FIG. 1).
  • Processor 605 may also be operatively coupled to a storage device 625. Storage device 625 may be any computer-operated hardware suitable for storing and/or retrieving data. In some aspects, storage device 625 may be integrated in server system 602. For example, server system 602 may include one or more hard disk drives as storage device 625. In other aspects, storage device 625 may be external to server system 602 and may be accessed by a plurality of server systems 602. For example, storage device 625 may include multiple storage units such as hard disks or solid-state disks in a redundant array of inexpensive disks (RAID) configuration. Storage device 625 may include a storage area network (SAN) and/or a network attached storage (NAS) system.
  • In some aspects, processor 605 may be operatively coupled to storage device 625 via a storage interface 620. Storage interface 620 may be any component capable of providing processor 605 with access to storage device 625. Storage interface 620 may include, for example, an Advanced Technology Attachment (ATA) adapter, a Serial ATA (SATA) adapter, a Small Computer System Interface (SCSI) adapter, a RAID controller, a SAN adapter, a network adapter, and/or any component providing processor 605 with access to storage device 625.
  • Memory areas 510 (shown in FIG. 3) and 610 may include, but are not limited to, random access memory (RAM) such as dynamic RAM (DRAM) or static RAM (SRAM), read-only memory (ROM), erasable programmable read-only memory (EPROM), electrically erasable programmable read-only memory (EEPROM), and non-volatile RAM (NVRAM). The above memory types are example only, and are thus not limiting as to the types of memory usable for storage of a computer program.
  • The computer systems and computer-implemented methods discussed herein may include additional, less, or alternate actions and/or functionalities, including those discussed elsewhere herein. The computer systems may include or be implemented via computer-executable instructions stored on non-transitory computer-readable media. The methods may be implemented via one or more local or remote processors, transceivers, servers, and/or sensors (such as processors, transceivers, servers, and/or sensors mounted on vehicle or mobile devices, or associated with smart infrastructure or remote servers), and/or via computer executable instructions stored on non-transitory computer-readable media or medium.
  • In some aspects, a computing device is configured to implement machine learning, such that the computing device “learns” to analyze, organize, and/or process data without being explicitly programmed. Machine learning may be implemented through machine learning (ML) methods and algorithms. In one aspect, a machine learning (ML) module is configured to implement ML methods and algorithms. In some aspects, ML methods and algorithms are applied to data inputs and generate machine learning (ML) outputs. Data inputs may further include: sequencing data, sensor data, image data, video data, telematics data, authentication data, authorization data, security data, mobile device data, geolocation information, transaction data, personal identification data, financial data, usage data, weather pattern data, “big data” sets, and/or user preference data. In some aspects, data inputs may include certain ML outputs.
  • In some aspects, at least one of a plurality of ML methods and algorithms may be applied, which may include but are not limited to: linear or logistic regression, instance-based algorithms, regularization algorithms, decision trees, Bayesian networks, cluster analysis, association rule learning, artificial neural networks, deep learning, dimensionality reduction, and support vector machines. In various aspects, the implemented ML methods and algorithms are directed toward at least one of a plurality of categorizations of machine learning, such as supervised learning, unsupervised learning, and reinforcement learning.
  • In one aspect, ML methods and algorithms are directed toward supervised learning, which involves identifying patterns in existing data to make predictions about subsequently received data. Specifically, ML methods and algorithms directed toward supervised learning are “trained” through training data, which includes example inputs and associated example outputs. Based on the training data, the ML methods and algorithms may generate a predictive function that maps outputs to inputs and utilize the predictive function to generate ML outputs based on data inputs. The example inputs and example outputs of the training data may include any of the data inputs or ML outputs described above.
  • In another aspect, ML methods and algorithms are directed toward unsupervised learning, which involves finding meaningful relationships in unorganized data. Unlike supervised learning, unsupervised learning does not involve user-initiated training based on example inputs with associated outputs. Rather, in unsupervised learning, unlabeled data, which may be any combination of data inputs and/or ML outputs as described above, is organized according to an algorithm-determined relationship.
  • In yet another aspect, ML methods and algorithms are directed toward reinforcement learning, which involves optimizing outputs based on feedback from a reward signal. Specifically ML methods and algorithms directed toward reinforcement learning may receive a user-defined reward signal definition, receive a data input, utilize a decision-making model to generate an ML output based on the data input, receive a reward signal based on the reward signal definition and the ML output, and alter the decision-making model so as to receive a stronger reward signal for subsequently generated ML outputs. The reward signal definition may be based on any of the data inputs or ML outputs described above. In one aspect, an ML module implements reinforcement learning in a user recommendation application. The ML module may utilize a decision-making model to generate a ranked list of options based on user information received from the user and may further receive selection data based on a user selection of one of the ranked options. A reward signal may be generated based on comparing the selection data to the ranking of the selected option. The ML module may update the decision-making model such that subsequently generated rankings more accurately predict a user selection.
  • As will be appreciated based upon the foregoing specification, the above-described aspects of the disclosure may be implemented using computer programming or engineering techniques including computer software, firmware, hardware or any combination or subset thereof. Any such resulting program, having computer-readable code means, may be embodied or provided within one or more computer-readable media, thereby making a computer program product, i.e., an article of manufacture, according to the discussed aspects of the disclosure. The computer-readable media may be, for example, but is not limited to, a fixed (hard) drive, diskette, optical disk, magnetic tape, semiconductor memory such as read-only memory (ROM), and/or any transmitting/receiving medium, such as the Internet or other communication network or link. The article of manufacture containing the computer code may be made and/or used by executing the code directly from one medium, by copying the code from one medium to another medium, or by transmitting the code over a network.
  • These computer programs (also known as programs, software, software applications, “apps”, or code) include machine instructions for a programmable processor, and can be implemented in a high-level procedural and/or object-oriented programming language, and/or in assembly/machine language. As used herein, the terms “machine-readable medium” “computer-readable medium” refers to any computer program product, apparatus and/or device (e.g., magnetic discs, optical disks, memory, Programmable Logic Devices (PLDs)) used to provide machine instructions and/or data to a programmable processor, including a machine-readable medium that receives machine instructions as a machine-readable signal. The “machine-readable medium” and “computer-readable medium,” however, do not include transitory signals. The term “machine-readable signal” refers to any signal used to provide machine instructions and/or data to a programmable processor.
  • As used herein, a processor may include any programmable system including systems using micro-controllers, reduced instruction set circuits (RISC), application specific integrated circuits (ASICs), logic circuits, and any other circuit or processor capable of executing the functions described herein. The above examples are examples only, and are thus not intended to limit in any way the definition and/or meaning of the term “processor.”
  • As used herein, the terms “software” and “firmware” are interchangeable, and include any computer program stored in memory for execution by a processor, including RAM memory, ROM memory, EPROM memory, EEPROM memory, and non-volatile RAM (NVRAM) memory. The above memory types are example only, and are thus not limiting as to the types of memory usable for storage of a computer program.
  • In one aspect, a computer program is provided, and the program is embodied on a computer-readable medium. In one aspect, the system is executed on a single computer system, without requiring a connection to a server computer. In a further aspect, the system is being run in a Windows® environment (Windows is a registered trademark of Microsoft Corporation, Redmond, Wash.). In yet another aspect, the system is run on a mainframe environment and a UNIX® server environment (UNIX is a registered trademark of X/Open Company Limited located in Reading, Berkshire, United Kingdom). The application is flexible and designed to run in various different environments without compromising any major functionality.
  • In some aspects, the system includes multiple components distributed among a plurality of computing devices. One or more components may be in the form of computer-executable instructions embodied in a computer-readable medium. The systems and processes are not limited to the specific aspects described herein. In addition, components of each system and each process can be practiced independent and separate from other components and processes described herein. Each component and process can also be used in combination with other assembly packages and processes. The present aspects may enhance the functionality and functioning of computers and/or computer systems.
  • Definitions and methods described herein are provided to better define the present disclosure and to guide those of ordinary skill in the art in the practice of the present disclosure. Unless otherwise noted, terms are to be understood according to conventional usage by those of ordinary skill in the relevant art.
  • In some embodiments, numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth, used to describe and claim certain embodiments of the present disclosure are to be understood as being modified in some instances by the term “about.” In some embodiments, the term “about” is used to indicate that a value includes the standard deviation of the mean for the device or method being employed to determine the value. In some embodiments, the numerical parameters set forth in the written description and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by a particular embodiment. In some embodiments, the numerical parameters should be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of some embodiments of the present disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as practicable. The numerical values presented in some embodiments of the present disclosure may contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. The recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. The recitation of discrete values is understood to include ranges between each value.
  • In some embodiments, the terms “a” and “an” and “the” and similar references used in the context of describing a particular embodiment (especially in the context of certain of the following claims) can be construed to cover both the singular and the plural, unless specifically noted otherwise. In some embodiments, the term “or” as used herein, including the claims, is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive.
  • The terms “comprise,” “have” and “include” are open-ended linking verbs. Any forms or tenses of one or more of these verbs, such as “comprises,” “comprising,” “has,” “having,” “includes” and “including,” are also open-ended. For example, any method that “comprises,” “has” or “includes” one or more steps is not limited to possessing only those one or more steps and can also cover other unlisted steps. Similarly, any composition or device that “comprises,” “has” or “includes” one or more features is not limited to possessing only those one or more features and can cover other unlisted features.
  • All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. “such as”) provided with respect to certain embodiments herein is intended merely to better illuminate the present disclosure and does not pose a limitation on the scope of the present disclosure otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the present disclosure.
  • Groupings of alternative elements or embodiments of the present disclosure disclosed herein are not to be construed as limitations. Each group member can be referred to and claimed individually or in any combination with other members of the group or other elements found herein. One or more members of a group can be included in, or deleted from, a group for reasons of convenience or patentability. When any such inclusion or deletion occurs, the specification is herein deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.
  • Any publications, patents, patent applications, and other references cited in this application are incorporated herein by reference in their entirety for all purposes to the same extent as if each individual publication, patent, patent application or other reference was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Citation of a reference herein shall not be construed as an admission that such is prior art to the present disclosure.
  • Having described the present disclosure in detail, it will be apparent that modifications, variations, and equivalent embodiments are possible without departing the scope of the present disclosure defined in the appended claims. Furthermore, it should be appreciated that all examples in the present disclosure are provided as non-limiting examples.
  • EXAMPLES
  • The following examples illustrate various aspects of the disclosure.
  • Example 1: Genome Sequencing as an Alternative to Cytogenetic Analysis in Myeloid Cancers
  • To develop and validate a whole-genome sequencing method for detecting genomic profiling in patients with acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS), the following experiments were conducted.
  • Patients
  • All the samples that were included in this study were obtained from patients with a known or suspected diagnosis of AML or MDS. All the patients provided written informed consent for genomic sequencing studies. Samples were selected for sequencing for three specific purposes: 1) WGS performance assessment, 2) Establishing diagnostic yield and clinical feasibility, and 3) Evaluation of risk prediction in patients with unsuccessful or incomplete cytogenetic studies. To achieve these objectives, a combination of retrospective and prospective patient cohorts was used as described below. Retrospective samples were obtained from cryopreserved diagnostic bone marrow or peripheral-blood specimens. Prospective samples were obtained from fresh bone marrow aspirate or peripheral-blood specimens collected from consecutive, unselected patients for whom clinical cytogenetic analysis by means of karyotyping had been requested.
  • Retrospective samples from AML and MDS patients (N=146) included DNA extracted from either cryopreserved bone marrow (N=133) or peripheral blood (N=13) specimens. For WGS performance evaluation, 111 samples were selected based on DNA availability and results from conventional cytogenetic studies in order to include a wide range of chromosomal abnormalities, including risk-defining translocations, copy number alterations (CNAs), and either a complex or normal karyotype. Separately, to determine whether WGS could be used to predict outcomes for patients with unknown cytogenetics, 35 retrospective samples were selected from patients treated with induction chemotherapy and for whom cytogenetics was unknown, unsuccessful, or inconclusive at diagnosis. Of note, samples with successful cytogenetic studies from the prospective cohort below were also used for WGS performance evaluation, and likewise, prospective samples with unsuccessful or inconclusive cytogenetics were used for the analysis of WGS-based risk prediction in patients with unknown cytogenetics.
  • Evaluation of the feasibility and diagnostic yield of WGS compared to standard testing used samples from a cohort of 117 prospective patients. These samples included bone marrow aspirate (N=116) or peripheral blood (N=1) specimens from 117 consecutive, unselected patients for whom clinical cytogenetic analysis via karyotyping was requested. The only selection criteria for these patients were patient consent and that there was sufficient remaining specimen left after standard cytogenetic analysis to be used for sequencing; some samples required the addition of RPMI based media to wash residual material out of the sodium heparin tubes prior to DNA extraction. This cohort included patients with both successful and unsuccessful cytogenetic studies, and therefore contributed to WGS performance evaluation and WGS-based risk prediction analysis for patients with unknown cytogenetics.
  • Whole Genome Sequencing
  • Tumor-only WGS was performed in a CLIA-licensed environment clinical sequencing laboratory; no normal tissue comparator was used for this assay in order to reduce time, complexity, and cost, and because the purpose is to identify clearly pathogenic somatic events. that generally do not require a germline control. All samples were accessioned into the MGI laboratory information management system (LIMS) upon receipt prior to DNA extraction (for prospective samples) or library preparation (for retrospective samples received as DNA). DNA from prospective peripheral blood or bone marrow aspirate specimens was extracted using 200 uL of material with the QIAamp DNA mini kit (Qiagen, Hilden, Germany) as detailed in the package insert, followed by quantification with the Qubit 1.0 fluorometer High Sensitivity dsDNA assay (ThermoFisher, Waltham, Mass.). Subsequent WGS procedures are described below.
  • We processed samples and performed sequencing to a target coverage depth of 60×. This analysis involved the identification of mutations in 40 genes, genomewide copy number alterations greater than 5 Mbp, and structural variants matching 612 recurrent structural alterations in myeloid cancers. Details regarding genetic identification and structural variants are provided in Tables 51 and S2 below. We used the results of whole-genome sequencing to assign patients to a genetic risk group through the same classification systems that are used for conventional analyses.
  • Library Preparation
  • WGS library preparation used the Nextera Flex library preparation kit (cat #20015804, Illumina, Inc, San Diego, Calif.) along with dual unique index library adapters (cat #20015881). This onbead tagmentation-based library construction method was selected because it is fast, simple, and automatable, and thus fits well in a clinical testing environment where training of laboratory staff and turnaround time are important considerations. For this study, library construction was performed in a single day in batches of 2 to 16 samples by individual laboratory staff who followed the protocol detailed in the package insert without modification. In general, 500 ng of input DNA was used for library construction, although as little as 35 ng was used when DNA amounts were limiting. Completed libraries were accessioned into the LIMS, then assessed for size using an Agilent 2100 Bioanalyzer with a DNA High Sensitivity chip (Agilent, Santa Clara, Calif.), and quantified via Qubit (ThermoFisher, Waltham, Mass.). Final libraries were optionally quantified further via qPCR (generally on a subsequent day) using Kapa SYBR Fast qPCR library quantification (Roche, Basel Switzerland), and then diluted to 1 nM for equimolar pooling prior to sequencing.
  • Sequencing
  • Sequencing was performed on NovaSeq 6000 sequencing instruments (Illumina) using either S1 or S4 flowcells and 2×150 sequencing chemistry. Retrospective samples were sequenced on S4 flowcells in pools of 16 (or pools of 4 samples on one S4 lane using the XP lane loader) and prospective ‘real-time’ sequencing used 51 flow cells in pools of 3 samples, which is designed to yield >133 Gbp of raw sequence and 60× genome coverage per sample. Flowcell loading and sequencing were performed as recommended by the manufacturer. Times for sequencing are 25 and 44 hours for 51 and S4 flowcells, respectively, and were documented in the MGI LIMS system.
  • Data Processing
  • Completed sequencing runs were processed into aligned CRAM files using the GRCh38 human reference genome via two approaches:
      • Local processing: For retrospective samples, instrument data were processed and demultiplexed into FASTQ files using bcl2fastq (Illumina) via the in-house LIMS. Data were then aligned via a local installation of the DRAGEN (version 3.5.7) hardware-accelerated sequence processing software suite using default alignment parameters. Processing time with this approach for S4 flowcells were generally 47 hours for sequencing data transfer and 12 hours for demultiplexing. Alignment times using DRAGEN ranged from 20-40 minutes depending on coverage.
      • BaseSpace processing: To speed the processing of prospective samples, data generated on S1 flowcells were streamed from the NovaSeq instrument to the cloud-based BaseSpace sequence analysis platform. This saved time at both the sequencing and data processing steps, owing to the shorter run time for S1 flowcells and ‘on-demand’ rapid data processing.
  • After the sequencing run completed, demultiplexing and FASTQ generation was automatically launched in BaseSpace. Data were then aligned via manual launching of the DRAGEN Germline (alignment only) BaseSpace App (version 3.2.8, see https://basespace.illumina.com/apps/6840834/DRAGEN-Germline-Pipeline), which completed in about the same amount of time as the local DRAGEN installation. We note that the manual step of launching DRAGEN can be automated via the BaseSpace API to further reduce turnaround time.
  • Alignments in CRAM format generated using both the in-house and cloud-based procedures were used as input for the variant analysis workflow described below.
  • Variant Analysis and Reporting
  • Tumor-only variant analysis used a custom analysis workflow (‘ChromoSeq’) specified in the WDL workflow language and executed using the Cromwell workflow engine1 in dockerized compute containers, which is available for public use as a custom application on BaseSpace (https://basespace.illumina.com/apps/6984978/Chromoseq—pending BaseSpace approval for public release). Analysis involves three components: CNA identification, SV identification, and gene-level variant identification. All three of these components are subject to targeted analysis and filtering to yield variants that may be clinically relevant.
  • CNA Identification
  • Cytogenetically evident CNAs greater than 5 Mbp, which are of a size potentially detectable by karyotype analysis, are identified via a read-depth approach using a previously published purity and subclone-aware Hidden Markov Model, ichorCNA2; https://github.com/GavinHaLab/ichorCNA). The input for this script is a file with read counts in 500,000 bp nonoverlapping windows across the genome, either generated using bedtools3 or outputted directly from the DRAGEN mapping software during alignment using the command: dragen -r Sobj->{dragenref}--fastq-list $fastqfile --fastq-list-sample-id $samplename --enable-cnv true --cnv-targetbed/staging/garza testing/reference/all sequences.fa.bed --cnv-interval-width 500000 --output-directory $cramout output-file-prefix $sample --output-format CRAM --enable-bam-indexing true --enable-duplicate-marking true. Binned read counts are supplied to ichorCNA for normalization for GC content, mappability, and using a ‘panel of normals’ normalization file generated from 20 normal karyotype cases per the instructions on the ichorCNA github repo. Outputs from ichorCNA were text-processed via a custom PERL script (available upon request) to retain CNAs >5 Mbp and converted to VCF format, and then combined with SV calls (below) for input into the ChromoSeq reporting script.
  • CNA abundance as a percentage was calculated using the equation:

  • Abundance=(2{circumflex over ( )}L2R−1.0)/((CN/2.0−1.0)))*100,
  • where L2R is the log 2 normalized coverage ratio vs. a panel of normals and CN is the estimated copy number for the event.
  • SV Identification
  • SV identification is performed with the break-end caller Manta and broken into two ‘tiers’. In the first tier, recurrrent and risk defining events are detected with a high sensitivity approach, and in the second tier, novel SVs are subject to more rigorous filtering. Manta is run directly from the aligned CRAM file in ‘tumor’ mode and with custom parameters to increase the sensitivity and limit calls to those that are at least 100 kbp in length to reduce the number of calls with unknown clinical significance. SVs are then filtered to identify translocations, deletions, duplications, and inversions that overlap a curated list of 612 recurrent and/or risk-defining SVs obtained from published sources, including the WHO and the Atlas of Genetics and Cytogenetics in Oncology (see Table S2). Genomic events where both ends overlap one of these recurrent SVs are reported as ‘top-level’ findings in ChromoSeq without additional filtering. Although the remaining SVs will rarely be clinically relevant, they could include patient-specific events or identify cytogenetically cryptic rearrangements involving genes relevant for AML or MDS. We therefore perform rigorous annotation and filtering using a custom PERL script (available upon request) and report the remaining high-quality novel events as secondary findings. The following criteria must be met to yield a passing call: 1) at least 2 ‘paired and 2 ‘split’ reads supporting the break-ends, 2) absence of an overlapping call from a large set of SVs identified from >17,795 human genomes5, 3) coverage depth of deletion or duplication call must be <0.8 or >1.3 compared to the background6, respectively, and 4) a defined breakpoint must be identified and the spanning contig generated by Manta must map back to the reported breakpoints. This procedure dramatically reduces the number of reported calls. For example, the mean number of raw Manta calls per case is >5,000; after filtering we reported a mean of 11 calls across all 263 cases in this study (including recurrent SVs). SVs are then converted to VCF format, combined with CAN calls (from above), and annotated with VEP7 using Ensembl version 90, prior to reporting with the ChromoSeq reporting script.
  • TABLE S2
    Recurrent SVs in AML and MDS
    Chrom1 Chrom2 Gene_Pair
    Start End Start End Gene1 Strand Gene2 Strand
    chr1 3069210 3438621 chr1 2228694 2310119 PRDM16_SKI + +
    chr1 110338505 110346677 chr22 40410280 40636685 RBM15_MRTFA +
    chr1 148808465 149032955 chr5 150113836 150155860 PDE4DIP_PDGFRB +
    chr1 154157317 154192058 chr5 150113836 150155860 TPM3_PDGFRB
    chr1 186311651 186375325 chr8 38411138 38468834 TPR_FGFR1
    chr1 221701423 221742176 chr1 3069210 3438621 DUSP10_PRDM16 +
    chr1 234604268 234609525 chr17 40309193 40356796 IRF2BP2_RARA +
    chr10 134464 254626 chr17 51177424 51260066 ZMYND11_MBTD1 +
    chr10 21524674 21743630 chr9 41890313 42129250 MLLT10_CNTNAP3B +
    chr10 21524674 21743630 chrX 41333347 41364472 MLLT10_DDX3X + +
    chr10 32009009 32056431 chr4 54229096 54298247 KIF5B_PDGFRA +
    chr10 59788762 59906656 chr5 150113836 150155860 CCDC6_PDGFRB
    chr10 74824935 75032284 chr16 3725053 3880726 KAT6B_CREBBP +
    chr10 94501433 94602098 chr8 2938098 4994972 HELLS_CSMD1 +
    chr10 101130504 101137789 chr7 142299010 142813287 TLX1_TRB + +
    chr10 102394109 102402529 chr6 117288299 117425855 NFKB2_ROS1 +
    chr11 925808 1012245 chr14 52004802 52069228 AP2A2_NID2 +
    chr11 3675009 3797792 chr11 3675009 3797792 NUP98_NUP98
    chr11 3675009 3797792 chr11 108665024 108940930 NUP98_DDX10 +
    chr11 3675009 3797792 chr11 118436463 118526832 NUP98_KMT2A +
    chr11 3675009 3797792 chr12 280128 389454 NUP98_KDM5A
    chr11 3675009 3797792 chr17 7235027 7239506 NUP98_PHF23
    chr11 3675009 3797792 chr2 176092890 176095938 NUP98_HOXD13 +
    chr11 3675009 3797792 chr2 176107285 176109588 NUP98_HOXD11 +
    chr11 3675009 3797792 chr20 41028817 41124487 NUP98_TOP1 +
    chr11 3675009 3797792 chr3 87259403 87276587 NUP98_POU1F1
    chr11 3675009 3797792 chr3 100401192 100456319 NUP98_LNP1 +
    chr11 3675009 3797792 chr5 177133078 177300210 NUP98_NSD1 +
    chr11 3675009 3797792 chr6 138773508 138793317 NUP98_CCDC28A +
    chr11 3675009 3797792 chr7 27162434 27165530 NUP98_HOXA9
    chr11 3675009 3797792 chr7 27181514 27185216 NUP98_HOXA11
    chr11 3675009 3797792 chr7 27194363 27200106 NUP98_HOXA13
    chr11 3675009 3797792 chr8 38269696 38382272 NUP98_NSD3
    chr11 3675009 3797792 chr9 15464065 15511019 NUP98_PSIP1
    chr11 3675009 3797792 chr9 129665640 129722674 NUP98_PRRX2 +
    chr11 3675009 3797792 chrX 150983285 150990775 NUP98_HMGB3 +
    chr11 3855701 4093209 chr5 177133078 177300210 STIM1_NSD1 + +
    chr11 8106092 8169043 chr7 142299010 142813287 RIC3_TRB +
    chr11 8106092 8169043 chr7 142801040 142802748 RIC3_TRBC2 +
    chr11 20156154 20160613 chr9 36833274 37034185 DBX1_PAX5
    chr11 34051682 34101156 chr5 150113836 150155860 CAPRIN1_PDGFRB +
    chr11 59142747 59155039 chr6 135181314 135219171 FAM111A_MYB + +
    chr11 62559600 62573973 chr6 73368554 73395093 EEF1G_OOEP
    chr11 72002864 72080693 chr17 40309193 40356796 NUMA1_RARA +
    chr11 72814405 72843674 chr11 118436463 118526832 ATG16L2_KMT2A + +
    chr11 85957687 86069097 chr10 21524674 21743630 PICALM_MLLT10 +
    chr11 101451563 101583928 chr8 109240918 109334385 TRPC6_NUDCD1
    chr11 114059592 114250676 chr17 40309193 40356796 ZBTB16_RARA + +
    chr11 117427772 117797261 chr11 118436463 118526832 DSCAML1_KMT2A +
    chr11 117836980 117877486 chr11 118436463 118526832 FXYD6_KMT2A +
    chr11 118436463 118526832 chr1 51354262 51519328 KMT2A_EPS15 +
    chr11 118436463 118526832 chr1 151057757 151068497 KMT2A_MLLT11 + +
    chr11 118436463 118526832 chr10 20783905 21174187 KMT2A_NEBL +
    chr11 118436463 118526832 chr10 21524674 21743630 KMT2A_MLLT10 + +
    chr11 118436463 118526832 chr10 26746595 26861087 KMT2A_ABI1 +
    chr11 118436463 118526832 chr10 68560655 68694482 KMT2A_TET1 + +
    chr11 118436463 118526832 chr11 73308288 73369091 KMT2A_ARHGEF17 + +
    chr11 118436463 118526832 chr11 85957687 86069097 KMT2A_PICALM +
    chr11 118436463 118526832 chr11 95976597 96343180 KMT2A_MAML2 +
    chr11 118436463 118526832 chr11 118436463 118526832 KMT2A_KMT2A + +
    chr11 118436463 118526832 chr11 119206275 119308149 KMT2A_CBL + +
    chr11 118436463 118526832 chr11 120337077 120489936 KMT2A_ARHGEF12 + +
    chr11 118436463 118526832 chr12 55757273 55817756 KMT2A_SARNP +
    chr11 118436463 118526832 chr14 66507406 67181803 KMT2A_GPHN + +
    chr11 118436463 118526832 chr14 104865279 104896742 KMT2A_CEP170B + +
    chr11 118436463 118526832 chr15 40594019 40664342 KMT2A_KNL1 + +
    chr11 118436463 118526832 chr15 40807088 40815084 KMT2A_ZFYVE19 + +
    chr11 118436463 118526832 chr16 3725053 3880726 KMT2A_CREBBP +
    chr11 118436463 118526832 chr17 9913849 10198551 KMT2A_GAS7 +
    chr11 118436463 118526832 chr17 38705541 38729803 KMT2A_MLLT6 + +
    chr11 118436463 118526832 chr17 38869858 38921770 KMT2A_LASP1 + +
    chr11 118436463 118526832 chr17 40309193 40356796 KMT2A_RARA + +
    chr11 118436463 118526832 chr19 4360369 4400547 KMT2A_SH3GL1 +
    chr11 118436463 118526832 chr19 6210378 6279948 KMT2A_MLLT1 +
    chr11 118436463 118526832 chr19 18442662 18522127 KMT2A_ELL +
    chr11 118436463 118526832 chr2 203328218 203447723 KMT2A_ABI2 + +
    chr11 118436463 118526832 chr22 41091785 41180079 KMT2A_EP300 + +
    chr11 118436463 118526832 chr3 48673843 48685927 KMT2A_NCKIPSD +
    chr11 118436463 118526832 chr3 108549868 108589452 KMT2A_CIP2A +
    chr11 118436463 118526832 chr3 155870535 155944026 KMT2A_GMPS + +
    chr11 118436463 118526832 chr3 188212932 188890671 KMT2A_LPP + +
    chr11 118436463 118526832 chr4 39822862 39977956 KMT2A_PDS5A +
    chr11 118436463 118526832 chr4 48497362 48780299 KMT2A_FRYL +
    chr11 118436463 118526832 chr4 52590971 52659335 KMT2A_USP46 +
    chr11 118436463 118526832 chr4 76949808 77040384 KMT2A_SEPT11 + +
    chr11 118436463 118526832 chr4 86935001 87141054 KMT2A_AFF1 + +
    chr11 118436463 118526832 chr4 185585443 185956368 KMT2A_SORBS2 +
    chr11 118436463 118526832 chr5 127517608 127555089 KMT2A_PRRC1 + +
    chr11 118436463 118526832 chr5 139274103 139331671 KMT2A_MATR3 + +
    chr11 118436463 118526832 chr5 142770376 143229011 KMT2A_ARHGAP26 + +
    chr11 118436463 118526832 chr5 160251656 160312928 KMT2A_CCNJL +
    chr11 118436463 118526832 chr6 70667775 70862015 KMT2A_SMAP1 + +
    chr11 118436463 118526832 chr6 89829899 89871412 KMT2A_CASP8AP2 + +
    chr11 118436463 118526832 chr6 108559834 108684774 KMT2A_FOXO3 + +
    chr11 118436463 118526832 chr6 136557046 136792518 KMT2A_MAP3K5 +
    chr11 118436463 118526832 chr6 167826990 167972020 KMT2A_AFDN + +
    chr11 118436463 118526832 chr7 5306789 5423546 KMT2A_TNRC18 +
    chr11 118436463 118526832 chr7 87628412 87832296 KMT2A_RUNDC3B + +
    chr11 118436463 118526832 chr9 20341664 20622543 KMT2A_MLLT3 +
    chr11 118436463 118526832 chr9 96450200 96491336 KMT2A_HABP4 + +
    chr11 118436463 118526832 chr9 121567101 121785528 KMT2A_DAB2IP + +
    chr11 118436463 118526832 chr9 129887186 130043194 KMT2A_FNBP1 +
    chr11 118436463 118526832 chr9 131009081 131093059 KMT2A_LAMC3 + +
    chr11 118436463 118526832 chrX 71096196 71103535 KMT2A_FOXO4 + +
    chr11 118436463 118526832 chrX 119615723 119693370 KMT2A_SEPT6 +
    chr11 118436463 118526832 chrX 135760156 135768191 KMT2A_CT45A3 +
    chr11 118436463 118526832 chrX 135811980 135820012 KMT2A_CT45A2 +
    chr11 118436463 118526832 chrX 154348525 154374638 KMT2A_FLNA +
    chr11 119206275 119308149 chr9 470290 746105 CBL_KANK1 + +
    chr11 122655674 122814473 chr7 23504779 23532041 UBASH3B_TRA2A +
    chr12 991207 1495933 chr5 150113836 150155860 ERC1_PDGFRB +
    chr12 6666476 6689510 chr5 64165881 64372869 ZNF384_RNF180 +
    chr12 10158300 10172138 chrX 123859811 123913976 OLR1_XIAP +
    chr12 10170541 10191785 chrX 123859811 123913976 TMEM52B_XIAP + +
    chr12 11649853 11895402 chr1 3069210 3438621 ETV6_PRDM16 + +
    chr12 11649853 11895402 chr1 179099326 179229601 ETV6_ABL2 +
    chr12 11649853 11895402 chr10 99396869 99431569 ETV6_GOT1 +
    chr12 11649853 11895402 chr12 11649853 11895402 ETV6_ETV6 + +
    chr12 11649853 11895402 chr12 56595595 56636366 ETV6_BAZ2A +
    chr12 11649853 11895402 chr12 70638081 70920843 ETV6_PTPRR +
    chr12 11649853 11895402 chr15 87859750 88256747 ETV6_NTRK3 +
    chr12 11649853 11895402 chr17 8144054 8156360 ETV6_PER1 +
    chr12 11649853 11895402 chr18 44680886 45068510 ETV6_SETBP1 + +
    chr12 11649853 11895402 chr3 41194740 41239949 ETV6_CTNNB1 + +
    chr12 11649853 11895402 chr3 169084760 169663470 ETV6_MECOM +
    chr12 11649853 11895402 chr4 54009788 54064690 ETV6_CHIC2 +
    chr12 11649853 11895402 chr4 54229096 54298247 ETV6_PDGFRA + +
    chr12 11649853 11895402 chr5 131949975 132011914 ETV6_ACSL6 +
    chr12 11649853 11895402 chr5 150113836 150155860 ETV6_PDGFRB +
    chr12 11649853 11895402 chr5 158695919 159099761 ETV6_EBF1 +
    chr12 11649853 11895402 chr6 115931148 116060758 ETV6_FRK +
    chr12 11649853 11895402 chr6 124962544 125092633 ETV6_RNF217 + +
    chr12 11649853 11895402 chr7 36389805 36453791 ETV6_ANLN + +
    chr12 11649853 11895402 chr7 36854360 37449249 ETV6_ELMO1 +
    chr12 11649853 11895402 chr8 55879834 56014168 ETV6_LYN + +
    chr12 11649853 11895402 chr8 98454694 98942571 ETV6_STK3 +
    chr12 11649853 11895402 chr9 4985244 5128183 ETV6_JAK2 + +
    chr12 11649853 11895402 chr9 90801786 90898549 ETV6_SYK + +
    chr12 11649853 11895402 chr9 130713945 130885683 ETV6_ABL1 + +
    chr12 14365631 14502935 chr9 4985244 5128183 ATF7IP_JAK2 + +
    chr12 26938382 26966553 chr8 38411138 38468834 FGFR1OP2_FGFR1 +
    chr12 48935722 48957551 chr6 149317533 149411395 ARF3_TAB2 +
    chr12 49018974 49055324 chr4 82819010 82900538 KMT2D_SEC31A
    chr12 51281281 51324668 chr5 150113836 150155860 BIN2_PDGFRB
    chr12 53452101 53481161 chr9 131394092 131500197 PCBP2_PRRC2B + +
    chr12 65824130 65966295 chr12 65824130 65966295 HMGA2_HMGA2 + +
    chr12 69239536 69274358 chr8 38411138 38468834 CPSF6_FGFR1 +
    chr12 71754873 71800285 chr12 71839717 71927248 RAB21_TBC1D15 + +
    chr12 104456970 104762014 chrX 120362084 120383249 CHST11_ATP1B4 + +
    chr12 108522579 108561389 chr5 150113836 150155860 SART3_PDGFRB
    chr12 109929801 109996389 chr5 150113836 150155860 GIT2_PDGFRB
    chr12 117453053 117968983 chr3 4303303 4317567 KSR2_SETMAR +
    chr12 121429098 121581015 chr5 132875378 132963634 KDM2B_AFF4
    chr12 121429098 121581015 chrY 13248378 13480673 KDM2B_UTY
    chr12 124324414 124535603 chr6 117288299 117425855 NCOR2_ROS1
    chr12 132489550 132585188 chr9 36833274 37034185 FBRSL1_PAX5 +
    chr13 19958669 20091829 chr8 38411138 38468834 ZMYM2_FGFR1 +
    chr13 48303725 48481890 chr7 15200317 15562015 RB1_AGMO +
    chr14 21621903 22552132 chr14 95709966 95714196 TRA_TCL1A +
    chr14 21621903 22552132 chr6 118460780 118710075 TRA_CEP85L +
    chr14 21621903 22552132 chr7 142299010 142813287 TRA_TRB + +
    chr14 21621903 22552132 chr9 21968104 21995301 TRA_CDKN2A +
    chr14 21621903 22552132 chr9 136494432 136545786 TRA_NOTCH1 +
    chr14 21621903 22552132 chrX 155065320 155147775 TRA_MTCP1 +
    chr14 22422545 22466577 chr14 95709966 95714196 TRD_TCL1A +
    chr14 22422545 22466577 chr5 170861869 171300015 TRD_RANBP17 + +
    chr14 22422545 22466577 chr5 171309283 171312134 TRD_TLX3 + +
    chr14 22422545 22466577 chr5 173232108 173235357 TRD_NKX2-5 +
    chr14 22422545 22466577 chr8 127736068 127741434 TRD_MYC + +
    chr14 22422545 22466577 chr8 127890627 128101253 TRD_PVT1 + +
    chr14 22422545 22466577 chr9 77716086 78031458 TRD_GNAQ +
    chr14 30893798 31026401 chr9 4985244 5128183 STRN3_JAK2 +
    chr14 50719762 50831121 chr5 150113836 150155860 NIN_PDGFRB
    chr14 73958009 74015928 chr6 135283531 135497745 ENTPD5_AHI1
    chr14 91271324 91417777 chr5 150113836 150155860 CCDC88C_PDGFRB
    chr14 91965990 92040059 chr5 150113836 150155860 TRIP11_PDGFRB
    chr14 99169286 99271485 chr14 99169286 99271485 BCL11B_BCL11B
    chr14 99169286 99271485 chr5 173232108 173235357 BCL11B_NKX2-5
    chr14 105586436 106879844 chr3 187721376 187745727 IGH_BCL6
    chr14 105586436 106879844 chr4 190173773 190175845 IGH_DUX4 +
    chr14 105586436 106879844 chr5 1253166 1295047 IGH_TERT
    chr14 105586436 106879844 chr5 112976735 113020970 IGH_DCP2 +
    chr14 105586436 106879844 chr5 158695919 159099761 IGH_EBF1
    chr14 105586436 106879844 chr6 391738 411447 IGH_IRF4 +
    chr14 105586436 106879844 chr7 55019100 55211628 IGH_EGFR +
    chr14 105586436 106879844 chr7 92468379 92477915 IGH_ERVW-1
    chr14 105586436 106879844 chr7 110663053 111562517 IGH_IMMP2L
    chr14 105586436 106879844 chr8 47736908 47739086 IGH_CEBPD
    chr14 105586436 106879844 chr9 36833274 37034185 IGH_PAX5
    chr14 105586436 106879844 chr9 124011609 124033301 IGH_LHX2 +
    chr15 43407208 43510614 chr5 150113836 150155860 TP53BP1_PDGFRB
    chr15 43510957 43531620 chr9 128947698 129007096 MAP1A_NUP188 + +
    chr15 73994728 74047812 chr17 40309193 40356796 PML_RARA + +
    chr15 75370932 75455783 chr9 33524393 33573001 SIN3A_ANKRD18B +
    chr15 80947328 80989828 chrX 41514933 41923169 MESD_CASK
    chr15 91853855 92172435 chr7 36854360 37449249 SLCO3A1_ELMO1 +
    chr16 176679 177522 chr5 150401669 150412929 HBA1_CD74 +
    chr16 3725053 3880726 chr7 142645960 142646467 CREBBP_TRBV23-1 +
    chr16 3725053 3880726 chr8 41929478 42052026 CREBBP_KAT6A
    chr16 10877197 10936388 chr9 5450502 5470566 CIITA_CD274 + +
    chr16 10877197 10936388 chr9 5510569 5571254 CIITA_PDCD1LG2 + +
    chr16 10877197 10936388 chr9 133097719 133149334 CIITA_RALGDS +
    chr16 11976737 12574289 chr9 4985244 5128183 SNX29_JAK2 + +
    chr16 15643266 15726353 chr5 150113836 150155860 NDE1_PDGFRB +
    chr16 31180109 31194871 chr21 38380029 38661780 FUS_ERG +
    chr16 31180109 31194871 chr9 128683654 128696400 FUS_SET + +
    chr16 67029146 67101058 chr16 15703171 15857011 CBFB_MYH11 +
    chr16 88874857 88977204 chr16 4314760 4339597 CBFA2T3_GLIS2 +
    chr17 5282299 5385812 chr5 150113836 150155860 RABEP1_PDGFRB +
    chr17 16029156 16215549 chr8 55879834 56014168 NCOR1_LYN +
    chr17 17042759 17192648 chr5 150113836 150155860 MPRIP_PDGFRB +
    chr17 20009362 20314138 chr5 150113836 150155860 SPECC1_PDGFRB +
    chr17 27456469 27626435 chrX 124375902 124963817 KSR1_TENM1 +
    chr17 29071123 29180412 chr5 150113836 150155860 MYO18A_PDGFRB
    chr17 29071123 29180412 chr8 38411138 38468834 MYO18A_FGFR1
    chr17 42199167 42276406 chr17 40309193 40356796 STAT5B_RARA +
    chr17 50183288 50201632 chr5 150113836 150155860 COL1A1_PDGFRB
    chr17 50962173 51120865 chr9 4985244 5128183 SPAG9_JAK2 +
    chr17 57256533 57684685 chr17 57256533 57684685 MSI2_MSI2 + +
    chr17 57256533 57684685 chr7 27162434 27165530 MSI2_HOXA9 +
    chr17 68515399 68551319 chr17 40309193 40356796 PRKAR1A_RARA + +
    chr17 80260867 80398786 chr8 127736068 127741434 RNF213_MYC + +
    chr17 82519714 82604607 chr7 74289474 74405943 FOXK2_CLIP2 + +
    chr18 2847029 2915993 chr9 36833274 37034185 EMILIN2_PAX5 +
    chr18 12785477 12884338 chr5 159263080 159286040 PTPN2_UBLCP1 +
    chr18 58671385 58754477 chr3 47850695 48088839 MALT1_MAP4 +
    chr19 2360237 2426239 chr9 36833274 37034185 TMPRSS9_PAX5 +
    chr19 10718092 10831884 chr7 93188585 93226524 DNM2_HEPACAM2 +
    chr19 13099032 13102867 chr7 142299010 142813287 LYL1_TRB +
    chr19 19385832 19508931 chr8 55879834 56014168 GATAD2A_LYN + +
    chr19 21020619 21060050 chr9 4985244 5128183 ZNF430_JAK2 + +
    chr19 34172565 34229515 chr9 130713945 130885683 LSM14A_ABL1 + +
    chr19 44748546 44760044 chr8 127736068 127741434 BCL3_MYC + +
    chr19 58183028 58213562 chr9 4985244 5128183 ZNF274_JAK2 + +
    chr19 58305318 58315663 chr8 38411138 38468834 ERVK3-1_FGFR1 +
    chr2 32946971 33399359 chr2 32357027 32618899 LTBP1_BIRC6 + +
    chr2 43230835 43596046 chr3 169084760 169663470 THADA_MECOM
    chr2 54456316 54671445 chr5 150113836 150155860 SPTBN1_PDGFRB +
    chr2 60450519 60554467 chr3 169084760 169663470 BCL11A_MECOM
    chr2 108719480 108785811 chr2 29192773 29921566 RANBP2_ALK +
    chr2 108719480 108785811 chr8 38411138 38468834 RANBP2_FGFR1 +
    chr2 108719480 108785811 chr9 130713945 130885683 RANBP2_ABL1 + +
    chr2 191678135 191696659 chr17 40309193 40356796 NABP1_RARA + +
    chr2 237627575 237780315 chr8 38411138 38468834 LRRFIP1_FGFR1 +
    chr20 18587892 18763917 chr5 150113836 150155860 DTD1_PDGFRB +
    chr20 32277663 32335011 chr7 142299010 142813287 KIF3B_TRB + +
    chr20 34714773 34825649 chr8 70051650 70071327 NCOA6_PRDM14
    chr20 44496223 44522085 chr9 37120538 37358149 SERINC3_ZCCHC7 +
    chr20 47209213 47356889 chr5 150113836 150155860 ZMYND8_PDGFRB
    chr20 52051662 52191779 chr21 34787800 35049344 ZFP64_RUNX1
    chr21 14961234 15065000 chr3 169084760 169663470 NRIP1_MECOM
    chr21 15730024 15880069 chr9 4985244 5128183 USP25_JAK2 + +
    chr21 29024628 29054488 chr21 34787800 35049344 USP16_RUNX1 +
    chr21 34516483 34615142 chr4 123396794 123403760 RCAN1_SPRY1 +
    chr21 34787800 35049344 chr1 3069210 3438621 RUNX1_PRDM16 +
    chr21 34787800 35049344 chr1 28736620 28769774 RUNX1_YTHDF2 +
    chr21 34787800 35049344 chr1 86424085 86456558 RUNX1_CLCA2 +
    chr21 34787800 35049344 chr1 151282311 151291903 RUNX1_ZNF687 +
    chr21 34787800 35049344 chr11 33542274 33674102 RUNX1_KIAA1549L +
    chr21 34787800 35049344 chr11 58526870 58578166 RUNX1_LPXN
    chr21 34787800 35049344 chr11 63998557 64166061 RUNX1_MACROD1
    chr21 34787800 35049344 chr16 88874857 88977204 RUNX1_CBFA2T3
    chr21 34787800 35049344 chr21 34787800 35049344 RUNX1_RUNX1
    chr21 34787800 35049344 chr3 169084760 169663470 RUNX1_MECOM
    chr21 34787800 35049344 chr3 169483670 169484080 RUNX1_RPL22P1
    chr21 34787800 35049344 chr4 151120286 151325632 RUNX1_SH3D19
    chr21 34787800 35049344 chr5 127517608 127555089 RUNX1_PRRC1 +
    chr21 34787800 35049344 chr5 129460264 129738683 RUNX1_ADAMTS19 +
    chr21 34787800 35049344 chr6 17582033 17582305 RUNX1_SUMO2P13 +
    chr21 34787800 35049344 chr7 6104883 6161564 RUNX1_USP42 +
    chr21 34787800 35049344 chr7 27242699 27247825 RUNX1_EVX1 +
    chr21 34787800 35049344 chr8 91954966 92103226 RUNX1_RUNX1T1
    chr21 34787800 35049344 chr8 105318691 105804532 RUNX1_ZFPM2 +
    chr21 34787800 35049344 chr8 115408495 115669001 RUNX1_TRPS1
    chr21 34787800 35049344 chrX 23667445 23686399 RUNX1_PRDX4 +
    chr21 38380029 38661780 chr4 190173773 190175845 ERG_DUX4 +
    chr21 42653749 42775509 chr8 85656441 85663039 PDE9A_REXO1L1P +
    chr22 22026075 22922913 chr4 1793292 1808872 IGL_FGFR3 + +
    chr22 22026075 22922913 chr6 391738 411447 IGL_IRF4 + +
    chr22 22026075 22922913 chr6 41934933 42050357 IGL_CCND3 +
    chr22 22026075 22922913 chr8 127890627 128101253 IGL_PVT1 + +
    chr22 23180209 23318037 chr4 54229096 54298247 BCR_PDGFRA + +
    chr22 23180209 23318037 chr8 38411138 38468834 BCR_FGFR1 +
    chr22 23180209 23318037 chr9 4985244 5128183 BCR_JAK2 + +
    chr22 23180209 23318037 chr9 126914773 127223164 BCR_RALGPS1 + +
    chr22 23180209 23318037 chr9 130713945 130885683 BCR_ABL1 + +
    chr22 41091785 41180079 chr7 27153715 27156677 EP300_HOXA7 +
    chr3 9397718 9478154 chr7 50304668 50405101 SETD5_IKZF1 + +
    chr3 10115591 10127190 chr3 10141007 10152220 BRK1_VHL + +
    chr3 12583600 12664226 chr3 12734706 12769457 RAF1_TMEM40
    chr3 15560703 15601852 chr3 15450132 15521751 HACL1_COLQ
    chr3 15667235 15859771 chr11 3675009 3797792 ANKRD28_NUP98
    chr3 16315855 16513706 chr8 127890627 128101253 RFTN1_PVT1 +
    chr3 28241594 28325142 chr3 27372720 27484420 CMC1_SLC4A7 +
    chr3 30606501 30694134 chr19 4044363 4066945 TGFBR2_ZBTB7A +
    chr3 37243176 37366751 chr5 150113836 150155860 GOLGA4_PDGFRB +
    chr3 37988058 38007188 chr9 137007233 137028922 VILL_ABCA2 +
    chr3 39051997 39096388 chr5 150113836 150155860 WDR48_PDGFRB +
    chr3 47016688 47163967 chr3 46921730 46982010 SETD2_CCDC12
    chr3 47585271 47781917 chr17 47970533 47981772 SMARCC1_CDK5RAP3 +
    chr3 47850695 48088839 chr18 58671385 58754477 MAP4_MALT1 +
    chr3 48599002 48610037 chr3 43365847 43622068 UQCRC1_ANO10
    chr3 48673843 48685927 chr3 48636468 48662915 NCKIPSD_CELSR3
    chr3 49940006 50077249 chr5 150053290 150113372 RBM6_CSF1R +
    chr3 100609588 100695479 chr3 100709360 100748942 ADGRG7_TFG + +
    chr3 100709360 100748942 chr3 100609588 100695479 TFG_ADGRG7 + +
    chr3 101324204 101513184 chr12 11649853 11895402 SENP7_ETV6 +
    chr3 121663202 121749767 chr13 28003273 28100592 GOLGB1_FLT3
    chr3 121663202 121749767 chr5 150113836 150155860 GOLGB1_PDGFRB
    chr3 128479426 128493185 chr3 169084760 169663470 GATA2_MECOM
    chr3 128479426 128493185 chr7 27162434 27165530 GATA2_HOXA9
    chr3 128479426 128493185 chr7 27171219 27180261 GATA2_HOXA10
    chr3 128571999 128576086 chr3 169084760 169663470 LINC01565_MECOM
    chr3 128620158 128681075 chr1 3069210 3438621 RPN1_PRDM16 +
    chr3 128620158 128681075 chr3 169084760 169663470 RPN1_MECOM
    chr3 134157132 134250744 chr21 33903452 33915980 RYK_ATP5PO
    chr3 136148921 136195846 chr3 177019354 177196478 MSL2_TBL1XR1
    chr3 152268039 152465779 chr9 36833274 37034185 MBNL1_PAX5 +
    chr3 152268039 152465779 chr9 130713945 130885683 MBNL1_ABL1 + +
    chr3 160494994 160565588 chr9 109640787 109946703 KPNA4_PALM2 +
    chr3 169084760 169663470 chr21 34787800 35049344 MECOM_RUNX1
    chr3 169084760 169663470 chr3 169084760 169663470 MECOM_MECOM
    chr3 169084760 169663470 chr7 92604920 92833917 MECOM_CDK6
    chr3 169084760 169663470 chr7 142299010 142813287 MECOM_TRB +
    chr3 172040553 172401665 chr3 169084760 169663470 FNDC3B_MECOM +
    chr3 177019354 177196478 chr17 40309193 40356796 TBL1XR1_RARA +
    chr3 177019354 177196478 chr3 139357405 139389732 TBL1XR1_COPB2
    chr3 177019354 177196478 chr3 189631426 189897279 TBL1XR1_TP63 +
    chr3 177019354 177196478 chr5 150053290 150113372 TBL1XR1_CSF1R
    chr3 180912301 180982753 chr3 177019354 177196478 FXR1_TBL1XR1 +
    chr3 186147200 186362237 chr4 15703095 15732787 DGKG_BST1 +
    chr3 187721376 187745727 chr13 46125919 46211348 BCL6_LCP1
    chr3 187721376 187745727 chr16 10877197 10936388 BCL6_CIITA +
    chr3 187721376 187745727 chr19 6677703 6720682 BCL6_C3
    chr3 187721376 187745727 chr3 152268039 152465779 BCL6_MBNL1 +
    chr3 187721376 187745727 chr6 37170202 37175426 BCL6_PIM1 +
    chr3 187721376 187745727 chr8 66562174 66613249 BCL6_MYBL1
    chr3 188212932 188890671 chr3 187721376 187745727 LPP_BCL6 +
    chr3 189631426 189897279 chr3 177019354 177196478 TP63_TBL1XR1 +
    chr4 1009978 1026891 chr4 1211447 1249137 FGFRL1_CTBP1 +
    chr4 1289850 1340137 chr4 1211447 1249137 MAEA_CTBP1 +
    chr4 1347315 1395989 chr4 1289850 1340137 UVSSA_MAEA + +
    chr4 26860690 27025381 chr7 50304668 50405101 STIM2_IKZF1 + +
    chr4 53377644 53459668 chr17 40309193 40356796 FIP1L1_RARA + +
    chr4 53377644 53459668 chr4 54229096 54298247 FIP1L1_PDGFRA + +
    chr4 54009788 54064690 chr12 11649853 11895402 CHIC2_ETV6 +
    chr4 54229096 54298247 chr10 91798311 91865276 PDGFRA_TNKS2 + +
    chr4 54229096 54298247 chr12 11649853 11895402 PDGFRA_ETV6 + +
    chr4 67468747 67545606 chr9 130713945 130885683 CENPC_ABL1 +
    chr4 78776377 78912185 chr12 6666476 6689510 BMP2K_ZNF384 +
    chr4 81087369 81215117 chr5 150113836 150155860 PRKG2_PDGFRB
    chr4 86935001 87141054 chr11 117427772 117797261 AFF1_DSCAML1 +
    chr4 86935001 87141054 chr11 117836980 117877486 AFF1_FXYD6 +
    chr4 86935001 87141054 chr11 118998141 119015745 AFF1_CCDC84 + +
    chr4 86935001 87141054 chr14 67819828 68730218 AFF1_RAD51B + +
    chr4 88592422 88708542 chr4 88709788 88730103 HERC3_FAM13A-AS1 + +
    chr4 108047544 108168956 chr6 42050521 42087461 LEF1_TAF8 +
    chr4 139716752 140154184 chr4 77157150 77170059 MAML3_CCNG2 +
    chr4 150264658 151015727 chr4 151120286 151325632 LRBA_SH3D19
    chr4 158124473 158173050 chr4 158210248 158255411 FAM198B_TMEM144 +
    chr4 190173773 190175845 chr10 133623894 133626792 DUX4_FRG2B +
    chr4 190173773 190175845 chr14 105586436 106879844 DUX4_IGH +
    chr4 190173773 190175845 chr21 38380029 38661780 DUX4_ERG +
    chr4 190173773 190175845 chr4 118467589 118554100 DUX4_CEP170P1 + +
    chr5 864272 892824 chr15 34343314 34357737 BRD9_NUTM1 +
    chr5 35856848 35879603 chr7 142801040 142802748 IL7R_TRBC2 + +
    chr5 36876758 37066413 chr12 11649853 11895402 NIPBL_ETV6 + +
    chr5 36876758 37066413 chr17 48621158 48626356 NIPBL_HOXB9 +
    chr5 36876758 37066413 chr7 27162434 27165530 NIPBL_HOXA9 +
    chr5 40759378 40798198 chr5 40512332 40755908 PRKAA1_TTC33
    chr5 55307759 55425581 chr11 116843401 117098421 MTREX_SIK3 +
    chr5 56815573 56896152 chr16 58157906 58197920 MAP3K1_CSNK2A2 +
    chr5 64165881 64372869 chr15 85380714 85749355 RNF180_AKAP13 + +
    chr5 65517765 65563171 chr11 118436463 118526832 CENPK_KMT2A +
    chr5 81413020 81751253 chr5 108747821 109196841 SSBP2_FER +
    chr5 81413020 81751253 chr5 150053290 150113372 SSBP2_CSF1R
    chr5 83940553 84384793 chr7 131110095 131487844 EDIL3_MKLN1 +
    chr5 88718240 88904257 chr11 118436463 118526832 MEF2C_KMT2A +
    chr5 98853984 98928957 chr21 34787800 35049344 CHD1_RUNX1
    chr5 112976735 113020970 chr14 61695539 61748258 DCP2_HIF1A + +
    chr5 122774995 122830108 chr9 130713945 130885683 SNX2_ABL1 + +
    chr5 132060528 132063204 chr14 105586436 106879844 IL3_IGH +
    chr5 134148934 134177038 chr5 132875378 132963634 SKP1_AFF4
    chr5 140691425 140699318 chr5 140700333 140706676 HARS2_ZMAT2 + +
    chr5 141639301 141651419 chr7 140719326 140924810 FCHSD1_BRAF
    chr5 144158158 144170659 chr5 143277930 143435512 YIPF5_NR3C1
    chr5 150113836 150155860 chr14 91271324 91417777 PDGFRB_CCDC88C
    chr5 150113836 150155860 chr14 91965990 92040059 PDGFRB_TRIP11
    chr5 150113836 150155860 chr17 29071123 29180412 PDGFRB_MYO18A
    chr5 150113836 150155860 chr20 18587892 18763917 PDGFRB_DTD1 +
    chr5 150401669 150412929 chr5 150113836 150155860 CD74_PDGFRB
    chr5 151029948 151087158 chr5 150113836 150155860 TNIP1_PDGFRB
    chr5 157785742 157859145 chr5 88718240 88904257 CLINT1_MEF2C
    chr5 158695919 159099761 chr9 4985244 5128183 EBF1_JAK2 +
    chr5 170861869 171300015 chr14 22422545 22466577 RANBP17_TRD + +
    chr5 171309283 171312134 chr14 99169286 99271485 TLX3_BCL11B +
    chr5 171387115 171410883 chr17 40309193 40356796 NPM1_RARA + +
    chr5 171387115 171410883 chr19 10350532 10380676 NPM1_TYK2 +
    chr5 171387115 171410883 chr3 158571162 158606460 NPM1_MLF1 + +
    chr5 172983756 173035445 chr12 62260337 62416389 ATP6V0E1_USP15 + +
    chr5 173056351 173139284 chr14 96392110 96489427 CREBRF_AK7 + +
    chr5 177133078 177300210 chr11 3675009 3797792 NSD1_NUP98 +
    chr5 177133078 177300210 chr11 61792636 61797244 NSD1_FEN1 + +
    chr5 177331561 177351852 chr14 96502372 96567111 LMAN2_PAPOLA +
    chr5 177331561 177351852 chr5 177133078 177300210 LMAN2_NSD1 +
    chr5 179614178 179624669 chr10 21524674 21743630 HNRNPH1_MLLT10 +
    chr5 179614178 179624669 chr21 38380029 38661780 HNRNPH1_ERG
    chr5 179820758 179838078 chr8 38411138 38468834 SQSTM1_FGFR1 +
    chr5 179820758 179838078 chr9 131125560 131234670 SQSTM1_NUP214 + +
    chr6 5998001 6007605 chr19 2511218 2702709 NRN1_GNG7
    chr6 17615034 17706834 chr9 130713945 130885683 NUP153_ABL1 +
    chr6 18223867 18264823 chr9 131125560 131234670 DEK_NUP214 +
    chr6 31676739 31680377 chr6 31686948 31703444 LY6G5C_ABHD16A
    chr6 33620364 33696574 chr6 36243202 36308595 ITPR3_PNPLA1 + +
    chr6 39299000 39314553 chr6 39329989 39725405 KCNK17_KIF6
    chr6 41934933 42050357 chr12 11649853 11895402 CCND3_ETV6 +
    chr6 44219504 44234151 chr6 44246165 44253888 SLC29A1_HSP90AB1 + +
    chr6 45898450 46080348 chr6 44828731 45377933 CLIC5_SUPT3H
    chr6 69675950 69867236 chr20 41402100 41618494 LMBRD1_CHD6
    chr6 75602046 75718278 chr6 123804140 124825657 SENP6_NKAIN2 + +
    chr6 85660949 85678748 chr6 85505495 85593913 SNHG5_SNX14
    chr6 89926528 90296908 chr11 19117128 19176415 BACH2_ZDHHC13 +
    chr6 106969830 106975465 chrX 48574523 48579066 CD24_RBM3 +
    chr6 115931148 116060758 chr12 11649853 11895402 FRK_ETV6 +
    chr6 124962544 125092633 chr12 11649853 11895402 RNF217_ETV6 + +
    chr6 130013698 130141449 chr6 127968778 128520674 L3MBTL3_PTPRK +
    chr6 135181314 135219171 chr16 89644430 89657845 MYB_CHMP1A +
    chr6 135181314 135219171 chr5 71455614 71567820 MYB_BDP1 + +
    chr6 135181314 135219171 chr6 135283531 135497745 MYB_AHI1 +
    chr6 135181314 135219171 chr6 143940301 144064599 MYB_PLAGL1 +
    chr6 135181314 135219171 chr7 156994050 157009075 MYB_MNX1 +
    chr6 135181314 135219171 chrX 48786553 48794308 MYB_GATA1 + +
    chr6 138773508 138793317 chr11 3675009 3797792 CCDC28A_NUP98 +
    chr6 143060853 143340290 chr17 30477361 30527592 AIG1_GOSR1 + +
    chr6 144291828 144853034 chr14 73958009 74015928 UTRN_ENTPD5 +
    chr6 156776019 157210779 chr12 6666476 6689510 ARID1B_ZNF384 +
    chr6 166999181 167052713 chr10 43077026 43130351 FGFR1OP_RET + +
    chr6 166999181 167052713 chr8 38411138 38468834 FGFR1OP_FGFR1 +
    chr6 167826990 167972020 chr11 118436463 118526832 AFDN_KMT2A + +
    chr7 876551 896434 chr7 5306789 5423546 GET4_TNRC18 +
    chr7 2631950 2664802 chr7 1815793 2233243 TTYH3_MAD1L1 +
    chr7 5190187 5233826 chr12 112013315 112023451 WIPI2_ERP29 + +
    chr7 6374522 6403977 chr6 75084325 75206051 RAC1_COL12A1 +
    chr7 10931950 10940256 chr7 12570685 12660179 NDUFA4_SCIN +
    chr7 16646130 16706523 chr7 65960683 65982314 BZW2_GUSB +
    chr7 27162434 27165530 chr8 107251511 107336522 HOXA9_ANGPT1
    chr7 27168898 27171915 chr6 109366513 109382812 HOXA10-AS_CD164 +
    chr7 27171219 27180261 chr19 1086578 1095357 HOXA10_POLR2E
    chr7 27171219 27180261 chr7 142299010 142813287 HOXA10_TRB +
    chr7 27181514 27185216 chr7 16646130 16706523 HOXA11_BZW2 +
    chr7 27184670 27189169 chr7 92604920 92833917 HOXA11-AS_CDK6 +
    chr7 27201843 27207259 chr3 18347939 18438773 HOTTIP_SATB1 +
    chr7 30289793 30289890 chr7 30284306 30367692 MIR550A1_ZNRF2 + +
    chr7 34928875 35038041 chr7 27181514 27185216 DPY19L1_HOXA11
    chr7 37683871 37741374 chr15 52756988 52790012 GPR141_ONECUT1 +
    chr7 38240023 38368055 chr14 95709966 95714196 TRG_TCL1A
    chr7 40126022 40134652 chr6 27866848 27867529 MPLKIP_HIST1H1B
    chr7 43112598 43566001 chr5 150113836 150155860 HECW1_PDGFRB +
    chr7 45736786 45765812 chr7 56011066 56051604 SEPT7P2_PSPH
    chr7 50304668 50405101 chr1 3069210 3438621 IKZF1_PRDM16 + +
    chr7 50304668 50405101 chr12 11649853 11895402 IKZF1_ETV6 + +
    chr7 50304668 50405101 chr12 55966768 55972784 IKZF1_CDK2 + +
    chr7 50304668 50405101 chr15 34343314 34357737 IKZF1_NUTM1 + +
    chr7 50304668 50405101 chr17 16415541 16437003 IKZF1_TRPV2 + +
    chr7 50304668 50405101 chr3 9397718 9478154 IKZF1_SETD5 + +
    chr7 50304668 50405101 chr7 48171459 48647495 IKZF1_ABCA13 + +
    chr7 50590062 50793462 chr7 3301447 4269000 GRB10_SDK1 +
    chr7 66114604 66154561 chr10 96593311 96720514 CRCP_PIK3AP1 +
    chr7 74657684 74760692 chr17 40309193 40356796 GTF2I_RARA + +
    chr7 75533299 75738947 chr5 150113836 150155860 HIP1_PDGFRB
    chr7 92560792 92590393 chr7 92604920 92833917 FAM133B_CDK6
    chr7 92604920 92833917 chr11 118436463 118526832 CDK6_KMT2A +
    chr7 92604920 92833917 chr14 36516416 36521149 CDK6_NKX2-1
    chr7 92604920 92833917 chr3 169084760 169663470 CDK6_MECOM
    chr7 92604920 92833917 chr5 171309283 171312134 CDK6_TLX3 +
    chr7 92604920 92833917 chr7 27269356 27269678 CDK6_RPL35P4
    chr7 99472892 99503650 chr7 99493239 99500297 ZNF789_ZNF394 +
    chr7 100852752 100867008 chr6 135181314 135219171 SLC12A9_MYB + +
    chr7 101815903 102283958 chr4 73436162 73456174 CUX1_AFP + +
    chr7 101815903 102283958 chr7 149126415 149182802 CUX1_ZNF398 + +
    chr7 101815903 102283958 chr8 38411138 38468834 CUX1_FGFR1 +
    chr7 107923817 108003255 chr7 116862472 116922049 LAMB1_CAPZA2 +
    chr7 116862472 116922049 chr19 58544468 58550716 CAPZA2_TRIM28 + +
    chr7 138460333 138589993 chr8 38411138 38468834 TRIM24_FGFR1 +
    chr7 139043519 139109648 chr13 42272152 42323267 ZC3HAV1_AKAP11 +
    chr7 142299010 142813287 chr10 101130504 101137789 TRB_TLX1 + +
    chr7 142299010 142813287 chr11 8224313 8268716 TRB_LMO1 +
    chr7 142299010 142813287 chr14 95709966 95714196 TRB_TCL1A +
    chr7 142299010 142813287 chr22 37125837 37149990 TRB_IL2RB +
    chr7 142299010 142813287 chr3 169084760 169663470 TRB_MECOM +
    chr7 142299010 142813287 chr7 27162434 27165530 TRB_HOXA9 +
    chr7 142299010 142813287 chr7 27181514 27185216 TRB_HOXA11 +
    chr7 142299010 142813287 chr8 127736068 127741434 TRB_MYC + +
    chr7 142299010 142813287 chrX 108732481 108736409 TRB_IRS4 +
    chr7 148697913 148800582 chr7 153887096 154894285 CUL1_DPP6 + +
    chr7 152134924 152436005 chr7 152759748 152855378 KMT2C_ACTR3B +
    chr7 156994050 157009075 chr12 11649853 11895402 MNX1_ETV6 +
    chr7 158730994 158829628 chr20 3471039 3651122 ESYT2_ATRN +
    chr8 17922856 18029944 chr9 4985244 5128183 PCM1_JAK2 + +
    chr8 23385782 23457695 chr8 18527300 19013686 ENTPD4_PSD3
    chr8 28890403 29053270 chr9 4985244 5128183 HMBOX1_JAK2 + +
    chr8 38411138 38468834 chr5 179820758 179838078 FGFR1_SQSTM1 +
    chr8 38411138 38468834 chr7 101815903 102283958 FGFR1_CUX1 +
    chr8 38411138 38468834 chr8 38411138 38468834 FGFR1_FGFR1
    chr8 41929478 42052026 chr16 3725053 3880726 KAT6A_CREBBP
    chr8 41929478 42052026 chr19 39776594 39786135 KAT6A_LEUTX +
    chr8 41929478 42052026 chr2 25733752 25878516 KAT6A_ASXL2
    chr8 41929478 42052026 chr20 47501901 47656877 KAT6A_NCOA3 +
    chr8 41929478 42052026 chr22 41091785 41180079 KAT6A_EP300 +
    chr8 41929478 42052026 chr8 41929478 42052026 KAT6A_KAT6A
    chr8 41929478 42052026 chr8 70109761 70403805 KAT6A_NCOA2
    chr8 42338454 42371808 chr8 132571952 132675617 POLB_LRRC6 +
    chr8 42896931 43030539 chr8 38411138 38468834 HOOK3_FGFR1 +
    chr8 51817574 51899186 chr7 549196 727341 PCMTD1_PRKAR1B
    chr8 56160903 56211279 chr7 142797455 142797502 PLAG1_TRBJ2-7 +
    chr8 99013265 99877580 chrX 123961313 124102656 VPS13B_STAG2 + +
    chr8 123219956 123241398 chr7 27184670 27189169 C8orf76_HOXA11-AS +
    chr8 127736068 127741434 chr6 44828731 45377933 MYC_SUPT3H +
    chr8 127736068 127741434 chr9 37120538 37358149 MYC_ZCCHC7 + +
    chr8 127736068 127741434 chr9 37438113 37465399 MYC_ZBTB5 +
    chr8 127890627 128101253 chr13 34942286 35672735 PVT1_NBEA + +
    chr8 127890627 128101253 chr3 169084760 169663470 PVT1_MECOM +
    chr8 127890627 128101253 chr8 125091852 125367120 PVT1_NSMCE2 + +
    chr8 127890627 128101253 chr8 130052106 130443660 PVT1_ASAP1 +
    chr8 127890627 128101253 chr9 37120538 37358149 PVT1_ZCCHC7 + +
    chr8 143833720 143840974 chr9 109015151 109119945 NRBP2_TMEM245
    chr9 470290 746105 chr5 150113836 150155860 KANK1_PDGFRB +
    chr9 2015185 2193620 chr12 6666476 6689510 SMARCA2_ZNF384 +
    chr9 2015185 2193620 chr9 2621833 2660053 SMARCA2_VLDLR + +
    chr9 3218301 3526001 chr9 4985244 5128183 RFX3_JAK2 +
    chr9 4985244 5128183 chr16 11976737 12574289 JAK2_SNX29 + +
    chr9 4985244 5128183 chr5 158695919 159099761 JAK2_EBF1 +
    chr9 15464065 15511019 chr11 3675009 3797792 PSIP1_NUP98
    chr9 21968104 21995301 chr14 21621903 22552132 CDKN2A_TRA +
    chr9 21968104 21995301 chr9 21455483 21456049 CDKN2A_IFNWP19 +
    chr9 21968104 21995301 chr9 21802648 21937651 CDKN2A_MTAP +
    chr9 33041763 33076659 chr9 4985244 5128183 SMU1_JAK2 +
    chr9 34179012 34252523 chr9 34086386 34126773 UBAP1_DCAF12 +
    chr9 35812959 35815021 chr9 35792153 35809732 HINT2_NPR2 +
    chr9 36833274 37034185 chr10 7818503 8016627 PAX5_TAF3 +
    chr9 36833274 37034185 chr11 33542274 33674102 PAX5_KIAA1549L +
    chr9 36833274 37034185 chr12 132489550 132585188 PAX5_FBRSL1 +
    chr9 36833274 37034185 chr14 76310711 76498475 PAX5_ESRRB +
    chr9 36833274 37034185 chr16 88874857 88977204 PAX5_CBFA2T3
    chr9 36833274 37034185 chr16 89720984 89740903 PAX5_ZNF276 +
    chr9 36833274 37034185 chr17 47896149 47928957 PAX5_SP2 +
    chr9 36833274 37034185 chr20 32277663 32335011 PAX5_KIF3B +
    chr9 36833274 37034185 chr20 32358329 32439319 PAX5_ASXL1 +
    chr9 36833274 37034185 chr20 32443061 32584890 PAX5_NOL4L
    chr9 36833274 37034185 chr20 46060984 46089952 PAX5_NCOA5
    chr9 36833274 37034185 chr3 152268039 152465779 PAX5_MBNL1 +
    chr9 36833274 37034185 chr6 10747830 10759774 PAX5_TMEM14B +
    chr9 36833274 37034185 chr7 86643913 86864884 PAX5_GRM3 +
    chr9 36833274 37034185 chr9 470290 746105 PAX5_KANK1 +
    chr9 36833274 37034185 chr9 20341664 20622543 PAX5_MLLT3
    chr9 36833274 37034185 chr9 36336403 36401198 PAX5_RNF38
    chr9 36833274 37034185 chr9 37120538 37358149 PAX5_ZCCHC7 +
    chr9 36833274 37034185 chr9 113165519 113221361 PAX5_FKBP15
    chr9 36833274 37034185 chrX 1462571 1537107 PAX5_P2RY8
    chr9 36833274 37034185 chrX 40051245 40177329 PAX5_BCOR
    chr9 36833274 37034185 chrX 120158560 120165630 PAX5_RHOXF2 +
    chr9 37120538 37358149 chr20 44496223 44522085 ZCCHC7_SERINC3 +
    chr9 37120538 37358149 chr9 36833274 37034185 ZCCHC7_PAX5 +
    chr9 37422665 37436990 chr19 6677703 6720682 GRHPR_C3 +
    chr9 37919133 38069211 chr11 118747765 118791136 SHB_DDX6
    chr9 71683365 71768884 chr8 27311481 27459390 CEMIP2_PTK2B +
    chr9 92711362 92764812 chr9 4985244 5128183 BICD2_JAK2 +
    chr9 95875700 95968840 chr19 41330322 41353911 ERCC6L2_TGFB1 +
    chr9 100302083 100352939 chr9 97674908 97697357 TEX10_XPA
    chr9 105662456 105663112 chr7 142299010 142813287 TAL2_TRB + +
    chr9 109640787 109946703 chr9 110048695 110172512 PALM2_AKAP2 + +
    chr9 111525158 111577844 chr9 125261831 125367207 ZNF483_GAPVD1 + +
    chr9 111896765 111935369 chr8 127890627 128101253 UGCG_PVT1 + +
    chr9 120388868 120580170 chr4 54229096 54298247 CDK5RAP2_PDGFRA +
    chr9 121075011 121177608 chr4 54657917 54740715 CNTRL_KIT + +
    chr9 121075011 121177608 chr8 38411138 38468834 CNTRL_FGFR1 +
    chr9 123379653 123930107 chr9 37120538 37358149 DENND1A_ZCCHC7 +
    chr9 124517274 124771277 chr9 133205279 133209250 NR6A1_OBP2B
    chr9 128683654 128696400 chr9 131125560 131234670 SET_NUP214 + +
    chr9 129887186 130043194 chr11 118436463 118526832 FNBP1_KMT2A +
    chr9 130713945 130885683 chr17 74748651 74769353 ABL1_SLC9A3R1 + +
    chr9 130713945 130885683 chr22 23180209 23318037 ABL1_BCR + +
    chr9 130713945 130885683 chr9 131394092 131500197 ABL1_PRRC2B + +
    chr9 131125560 131234670 chr22 16783411 16821699 NUP214_XKR3 +
    chr9 131125560 131234670 chr7 6374522 6403977 NUP214_RAC1 + +
    chr9 131125560 131234670 chr9 130713945 130885683 NUP214_ABL1 + +
    chr9 131394092 131500197 chr10 129467183 129768007 PRRC2B_MGMT + +
    chr9 131394092 131500197 chr22 23180209 23318037 PRRC2B_BCR + +
    chr9 136440096 136483759 chr9 136494432 136545786 SEC16A_NOTCH1
    chr9 136494432 136545786 chr9 136862118 136866286 NOTCH1_EDF1
    chr9 136658855 136672678 chr17 83079690 83095119 EGFL7_METRNL + +
    chr9 136862118 136866286 chr9 136494432 136545786 EDF1_NOTCH1
    chrX 1462571 1537107 chr9 36833274 37034185 P2RY8_PAX5
    chrX 2691132 2741309 chr9 4985244 5128183 CD99_JAK2 + +
    chrX 13734744 13769353 chr9 4985244 5128183 OFD1_JAK2 + +
    chrX 40051245 40177329 chr17 40309193 40356796 BCOR_RARA +
    chrX 49028730 49043486 chrX 71283191 71301166 TFE3_NONO +
    chrX 55075062 55078909 chrX 55009054 55031064 PAGE2B_ALAS2 +
    chrX 71118555 71142453 chr7 27162434 27165530 MED12_HOXA9 +
    chrX 71283191 71301166 chr2 43222401 43226609 NONO_ZFP36L2 +
    chrX 71283191 71301166 chrX 49028730 49043486 NONO_TFE3 +
    chrX 100969710 100990806 chrX 101009345 101052116 ARL13A_TRMT2B +
    chrX 123859811 123913976 chr12 10158300 10172138 XIAP_OLR1 +
    chrX 123961313 124102656 chr7 16646130 16706523 STAG2_BZW2 + +
    chrX 123961313 124102656 chrX 77504877 77786216 STAG2_ATRX +
    chrX 123961313 124102656 chrX 130384439 130385447 STAG2_GPR119 +
    chrX 130064873 130110716 chr21 38380029 38661780 ELF4_ERG
    chrX 138632677 139205023 chr6 134169245 134318058 FGF13_SGK1
  • Gene-Level Variant Identification
  • Gene mutations are identified in—85 kbp targeting 40 genes and gene hotspots that are recurrently mutated in AML or MDS8. This target space was selected to be identical to that of the targeted gene panel used for clinical testing of these patients at our institution, and is relatively small so that rare inherited variants (i.e., variants of uncertain significance (VUS)), are minimized. The primary variant caller is Varscan2, which is run in SNV and indel mode using custom parameters to enhance sensitivity. The indel caller Pindel and Manta are also run on exons 13-15 of FLT3 to identify FLT3 ITD alleles. In addition, a read count based ‘hotspot’ analysis is performed on 66 recurrently mutated positions to recover low abundance variants that are not detected by Varscan2 (a minimum variant read count of 3 is required to report these hotspot positions). Variant calls identified via these approaches are merged and harmonized using a custom python script (available upon request), and annotated with VEP using Ensembl version 90 prior to reporting.
  • Report Generation
  • Annotated CNA, SV, and gene mutation calls are combined with coverage QC information to generate a final text report using a custom python script (available upon request). This report includes the CNAs, recurrent SVs, and gene mutations identified by the above steps as ‘toplevel’ results. The remaining SVs that remained after filtering are reported in two categories. The first includes high-quality novel SVs that affect (overlap) a gene that is included in either the recurrent SV or gene mutation target space. The second category is all other high-quality novel SVs. Additional coverage QC metrics are also reported. This text file is copied to the final case directory along with data files (CRAM, and VCF) and graphical coverage plots from ichorCNA. The final text report is also used to generate a graphical ChromoSeq report, as shown in FIG. 9.
  • WGS Analysis for Study Patients
  • All retrospective samples were sequenced on S4 flowcells and processed using in-house demultiplexing, aligned with the local DRAGEN server, and analyzed on a local compute cluster with the ChromoSeq workflow. Prospective samples were sequenced on S1 flowcells and initially analyzed using the cloud-based approach on BaseSpace to record failure rates and turnaround times. ChromoSeq reports with QC metrics and variants for the prospective patients were reviewed in 1 hour sessions by board-certified molecular pathologists and a board-certified cytogeneticist and molecular geneticist without prior knowledge of the results from conventional testing. Exact times for processing steps shown in FIG. 7A were obtained from the MGI LIMS system and from the timestamp in the ChromoSeq text report. A final ChromoSeq analysis was performed on all prospective samples at the end of the study to harmonize the results and file formats (which changed over the course of the study) with the outputs from the retrospective samples.
  • Conventional Cytogenetic and Molecular Analysis
  • All cytogenetic and FISH analyses were performed according to standard clinical protocols. We obtained data regarding genetic mutations as part of standard diagnostic testing using polymerase-chain-reaction (PCR)—based assays for the internal tandem duplication mutation in FLT3 (FLT3-ITD) and the NPM1c mutation, a laboratory-developed clinical sequencing assay, or both. Cytogenetic and molecular results were used to assign patients to established European Leukemia Network (ELN) or IPSS-R risk categories.
  • Culture of cells from bone marrow or leukemic peripheral blood samples was performed per standard clinical protocols, followed by harvest, slide dropping, G-banding with trypsin/Wright stain, and analysis. Cytogenetic events were considered clonal if they occurred in at least two metaphases (at least three metaphases for monosomies). For the purposes of this study, cytogenetic analysis was called ‘unsuccessful’ if no metaphases were obtained for analysis, and ‘inconclusive’ if fewer than 20 metaphases were analyzed without detection of clonal abnormalities, which is similar to approaches taken by other studies. FISH results used for risk stratification and calculation of the yield of WGS in the prospective cohort were obtained from clinical reports performed at diagnosis. For AML patients, most FISH studies (60 of 68 patients) included the ELN-recommended panel of PML-RARA (LSI PML/RARA Dual Color, Dual Fusion, Abbot/Vysis) CBFB-MYH11 (LSI CBFB Dual Color, Break Apart Rearrangement Probe, Abbot/Vysis), RUNX1-RUNX1T1 (LSI RUNX1T1/RUNX1 Dual Color, Dual Fusion, Vysis), del(5q) (D5S630/D5S2064 Dual Color Probe, Cytocell/Aquarius), and del(7q) (LSI D7S486/D7Z1 Dual Color Probe, Abbot/Vysis). Additional FISH assays were also performed to confirm WGS findings but were not used to for risk group assignments.
  • Gene mutations were obtained as part of standard diagnostic testing using a commercially available PCR-based assay for the FLT3 internal tandem duplication mutation (ITD) (Invivoscribe, San Diego, Calif.), in-house testing for the NPM1c mutation, and/or a laboratory-developed clinical sequencing assay, including either clinical tumor/normal exome sequencing or a clinical gene panel that targets 40 recurrently mutated genes or gene hotspots in AML and MDS (Myeloseq; Department of Pathology and Immunology, Washington University School of Medicine, see Table
  • TABLE S1
    Target list for gene mutation identification
    Chrom Start End Exon Strand Gene Name GeneID TranscriptID
    chr1 114713797 114713981 NRAS_exon_3 NRAS ENSG00000213281 ENST00000369535
    chr1 114716047 114716163 NRAS_exon_2 NRAS ENSG00000213281 ENST00000369535
    chr1 36466356 36466911 CSF3R_exon_17 CSF3R ENSG00000119535 ENST00000373103
    chr1 36467226 36467314 CSF3R_exon_16 CSF3R ENSG00000119535 ENST00000373103
    chr1 36467554 36467654 CSF3R_exon_15 CSF3R ENSG00000119535 ENST00000373103
    chr1 36467818 36467965 CSF3R_exon_14 CSF3R ENSG00000119535 ENST00000373103
    chr1 36468071 36468224 CSF3R_exon_13 CSF3R ENSG00000119535 ENST00000373103
    chr1 36469152 36469260 CSF3R_exon_12 CSF3R ENSG00000119535 ENST00000373103
    chr1 36469648 36469843 CSF3R_exon_11 CSF3R ENSG00000119535 ENST00000373103
    chr1 36471429 36471649 CSF3R_exon_10 CSF3R ENSG00000119535 ENST00000373103
    chr1 36472062 36472142 CSF3R_exon_9 CSF3R ENSG00000119535 ENST00000373103
    chr1 36472234 36472394 CSF3R_exon_8 CSF3R ENSG00000119535 ENST00000373103
    chr1 36472513 36472689 CSF3R_exon_7 CSF3R ENSG00000119535 ENST00000373103
    chr1 36473431 36473625 CSF3R_exon_6 CSF3R ENSG00000119535 ENST00000373103
    chr1 36473760 36473890 CSF3R_exon_5 CSF3R ENSG00000119535 ENST00000373103
    chr1 36475373 36475676 CSF3R_exon_4 CSF3R ENSG00000119535 ENST00000373103
    chr1 36479429 36479499 CSF3R_exon_3 CSF3R ENSG00000119535 ENST00000373103
    chr1 43349260 43349362 MPL_exon_10 + MPL ENSG00000117400 ENST00000372470
    chr10 110567813 110567834 SMC3_exon_1 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110568934 110569016 SMC3_exon_2 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110573703 110573748 SMC3_exon_3 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110575332 110575406 SMC3_exon_4 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110577417 110577495 SMC3_exon_5 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110577831 110577917 SMC3_exon_6 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110578624 110578709 SMC3_exon_7 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110580900 110581024 SMC3_exon_8 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110581919 110582101 SMC3_exon_9 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110582558 110582645 SMC3_exon_10 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110583380 110583551 SMC3_exon_11 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110583837 110583965 SMC3_exon_12 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110584179 110584399 SMC3_exon_13 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110589601 110589711 SMC3_exon_14 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110589888 110589994 SMC3_exon_15 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110590408 110590575 SMC3_exon_16 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110590987 110591135 SMC3_exon_17 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110593069 110593226 SMC3_exon_18 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110596394 110596553 SMC3_exon_19 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110598135 110598293 SMC3_exon_20 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110599650 110599815 SMC3_exon_21 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110600435 110600549 SMC3_exon_22 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110601018 110601133 SMC3_exon_23 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110601633 110601887 SMC3_exon_24 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110601962 110602181 SMC3_exon_25 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110602470 110602668 SMC3_exon_26 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110602821 110603005 SMC3_exon_27 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110603180 110603293 SMC3_exon_28 + SMC3 ENSG00000108055 ENST00000361804
    chr10 110604227 110604302 SMC3_exon_29 + SMC3 ENSG00000108055 ENST00000361804
    chr11 119278163 119278300 CBL_exon_8 + CBL ENSG00000110395 ENST00000264033
    chr11 119278507 119278716 CBL_exon_9 + CBL ENSG00000110395 ENST00000264033
    chr11 32389057 32389182 WT1_exon_10 WT1 ENSG00000184937 ENST00000332351
    chr11 32391968 32392067 WT1_exon_9 WT1 ENSG00000184937 ENST00000332351
    chr11 32392662 32392758 WT1_exon_8 WT1 ENSG00000184937 ENST00000332351
    chr11 32396253 32396410 WT1_exon_7 WT1 ENSG00000184937 ENST00000332351
    chr11 32399944 32400047 WT1_exon_6 WT1 ENSG00000184937 ENST00000332351
    chr11 32416486 32416543 WT1_exon_5 WT1 ENSG00000184937 ENST00000332351
    chr11 32417573 32417657 WT1_exon_4 WT1 ENSG00000184937 ENST00000332351
    chr11 32427952 32428061 WT1_exon_3 WT1 ENSG00000184937 ENST00000332351
    chr11 32428493 32428622 WT1_exon_2 WT1 ENSG00000184937 ENST00000332351
    chr11 32434696 32435348 WT1_exon_1 WT1 ENSG00000184937 ENST00000332351
    chr12 112450315 112450515 PTPN11_exon_3 + PTPN11 ENSG00000179295 ENST00000351677
    chr12 112489021 112489178 PTPN11_exon_13 + PTPN11 ENSG00000179295 ENST00000351677
    chr12 112502141 112502259 PTPN11_exon_14 + PTPN11 ENSG00000179295 ENST00000351677
    chr12 11650124 11650163 ETV6_exon_1 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11752446 11752582 ETV6_exon_2 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11839136 11839307 ETV6_exon_3 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11853423 11853564 ETV6_exon_4 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11869420 11869972 ETV6_exon_5 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11884441 11884590 ETV6_exon_6 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11885922 11886029 ETV6_exon_7 + ETV6 ENSG00000139083 ENST00000396373
    chr12 11890937 11891046 ETV6_exon_8 + ETV6 ENSG00000139083 ENST00000396373
    chr12 25227231 25227415 KRAS_exon_3 KRAS ENSG00000133703 ENST00000256078
    chr12 25245271 25245387 KRAS_exon_2 KRAS ENSG00000133703 ENST00000256078
    chr13 28018463 28018592 FLT3_exon_20 FLT3 ENSG00000122025 ENST00000241453
    chr13 28033884 28033994 FLT3_exon_15 FLT3 ENSG00000122025 ENST00000241453
    chr13 28034079 28034217 FLT3_exon_14 FLT3 ENSG00000122025 ENST00000241453
    chr13 28034298 28034410 FLT3_exon_13 FLT3 ENSG00000122025 ENST00000241453
    chr15 90088584 90088750 IDH2_exon_4 IDH2 ENSG00000182054 ENST00000330062
    chr17 31095306 31095372 NF1_exon_1 + NF1 ENSG00000196712 ENST00000358273
    chr17 31155979 31156129 NF1_exon_2 + NF1 ENSG00000196712 ENST00000358273
    chr17 31159006 31159096 NF1_exon_3 + NF1 ENSG00000196712 ENST00000358273
    chr17 31163182 31163379 NF1_exon_4 + NF1 ENSG00000196712 ENST00000358273
    chr17 31169887 31170000 NF1_exon_5 + NF1 ENSG00000196712 ENST00000358273
    chr17 31181418 31181492 NF1_exon_6 + NF1 ENSG00000196712 ENST00000358273
    chr17 31181706 31181788 NF1_exon_7 + NF1 ENSG00000196712 ENST00000358273
    chr17 31182504 31182668 NF1_exon_8 + NF1 ENSG00000196712 ENST00000358273
    chr17 31200418 31200598 NF1_exon_9 + NF1 ENSG00000196712 ENST00000358273
    chr17 31201033 31201162 NF1_exon_10 + NF1 ENSG00000196712 ENST00000358273
    chr17 31201407 31201488 NF1_exon_11 + NF1 ENSG00000196712 ENST00000358273
    chr17 31206236 31206374 NF1_exon_12 + NF1 ENSG00000196712 ENST00000358273
    chr17 31214447 31214588 NF1_exon_13 + NF1 ENSG00000196712 ENST00000358273
    chr17 31219001 31219121 NF1_exon_14 + NF1 ENSG00000196712 ENST00000358273
    chr17 31221846 31221932 NF1_exon_15 + NF1 ENSG00000196712 ENST00000358273
    chr17 31223440 31223570 NF1_exon_16 + NF1 ENSG00000196712 ENST00000358273
    chr17 31225091 31225253 NF1_exon_17 + NF1 ENSG00000196712 ENST00000358273
    chr17 31226431 31226687 NF1_exon_18 + NF1 ENSG00000196712 ENST00000358273
    chr17 31227214 31227294 NF1_exon_19 + NF1 ENSG00000196712 ENST00000358273
    chr17 31227519 31227609 NF1_exon_20 + NF1 ENSG00000196712 ENST00000358273
    chr17 31229021 31229468 NF1_exon_21 + NF1 ENSG00000196712 ENST00000358273
    chr17 31229831 31229977 NF1_exon_22 + NF1 ENSG00000196712 ENST00000358273
    chr17 31230256 31230385 NF1_exon_23 + NF1 ENSG00000196712 ENST00000358273
    chr17 31230838 31230928 NF1_exon_24 + NF1 ENSG00000196712 ENST00000358273
    chr17 31232069 31232192 NF1_exon_25 + NF1 ENSG00000196712 ENST00000358273
    chr17 31232696 31232884 NF1_exon_26 + NF1 ENSG00000196712 ENST00000358273
    chr17 31232998 31233216 NF1_exon_27 + NF1 ENSG00000196712 ENST00000358273
    chr17 31235607 31235775 NF1_exon_28 + NF1 ENSG00000196712 ENST00000358273
    chr17 31235914 31236024 NF1_exon_29 + NF1 ENSG00000196712 ENST00000358273
    chr17 31248980 31249122 NF1_exon_30 + NF1 ENSG00000196712 ENST00000358273
    chr17 31252934 31253003 NF1_exon_31 + NF1 ENSG00000196712 ENST00000358273
    chr17 31258340 31258505 NF1_exon_32 + NF1 ENSG00000196712 ENST00000358273
    chr17 31259028 31259132 NF1_exon_33 + NF1 ENSG00000196712 ENST00000358273
    chr17 31260365 31260518 NF1_exon_34 + NF1 ENSG00000196712 ENST00000358273
    chr17 31261707 31261860 NF1_exon_35 + NF1 ENSG00000196712 ENST00000358273
    chr17 31265225 31265342 NF1_exon_36 + NF1 ENSG00000196712 ENST00000358273
    chr17 31325816 31326255 NF1_exon_37 + NF1 ENSG00000196712 ENST00000358273
    chr17 31327495 31327842 NF1_exon_38 + NF1 ENSG00000196712 ENST00000358273
    chr17 31330292 31330501 NF1_exon_39 + NF1 ENSG00000196712 ENST00000358273
    chr17 31334834 31335034 NF1_exon_40 + NF1 ENSG00000196712 ENST00000358273
    chr17 31336329 31336476 NF1_exon_41 + NF1 ENSG00000196712 ENST00000358273
    chr17 31336631 31336917 NF1_exon_42 + NF1 ENSG00000196712 ENST00000358273
    chr17 31337364 31337585 NF1_exon_43 + NF1 ENSG00000196712 ENST00000358273
    chr17 31337815 31337883 NF1_exon_44 + NF1 ENSG00000196712 ENST00000358273
    chr17 31338021 31338142 NF1_exon_45 + NF1 ENSG00000196712 ENST00000358273
    chr17 31338700 31338808 NF1_exon_46 + NF1 ENSG00000196712 ENST00000358273
    chr17 31340501 31340648 NF1_exon_47 + NF1 ENSG00000196712 ENST00000358273
    chr17 31343005 31343138 NF1_exon_48 + NF1 ENSG00000196712 ENST00000358273
    chr17 31349116 31349254 NF1_exon_49 + NF1 ENSG00000196712 ENST00000358273
    chr17 31350179 31350321 NF1_exon_50 + NF1 ENSG00000196712 ENST00000358273
    chr17 31352253 31352417 NF1_exon_51 + NF1 ENSG00000196712 ENST00000358273
    chr17 31356456 31356585 NF1_exon_52 + NF1 ENSG00000196712 ENST00000358273
    chr17 31356956 31357093 NF1_exon_53 + NF1 ENSG00000196712 ENST00000358273
    chr17 31357265 31357372 NF1_exon_54 + NF1 ENSG00000196712 ENST00000358273
    chr17 31358476 31358625 NF1_exon_55 + NF1 ENSG00000196712 ENST00000358273
    chr17 31358965 31359018 NF1_exon_56 + NF1 ENSG00000196712 ENST00000358273
    chr17 31360483 31360706 NF1_exon_57 + NF1 ENSG00000196712 ENST00000358273
    chr17 31374009 31374155 NF1_exon_58 + NF1 ENSG00000196712 ENST00000358273
    chr17 31937243 31937523 SUZ12_exon_1 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31940282 31940335 SUZ12_exon_2 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31940418 31940489 SUZ12_exon_3 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31947613 31947688 SUZ12_exon_4 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31966143 31966199 SUZ12_exon_5 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31973142 31973234 SUZ12_exon_6 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31975478 31975716 SUZ12_exon_7 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31976517 31976617 SUZ12_exon_8 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31982995 31983107 SUZ12_exon_9 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31988316 31988500 SUZ12_exon_10 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31993238 31993336 SUZ12_exon_11 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31993861 31994011 SUZ12_exon_12 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31994560 31994724 SUZ12_exon_13 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31995560 31995765 SUZ12_exon_14 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31996794 31996880 SUZ12_exon_15 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 31998654 31999003 SUZ12_exon_16 + SUZ12 ENSG00000178691 ENST00000322652
    chr17 60662991 60663552 PPM1D_exon_6 + PPM1D ENSG00000170836 ENST00000305921
    chr17 7669608 7669693 TP53_exon_11 TP53 ENSG00000141510 ENST00000269305
    chr17 7670605 7670718 TP53_exon_10 TP53 ENSG00000141510 ENST00000269305
    chr17 7673531 7673611 TP53_exon_9 TP53 ENSG00000141510 ENST00000269305
    chr17 76736796 76737163 SRSF2_exon_1 SRSF2 ENSG00000161547 ENST00000392485
    chr17 7673697 7673840 TP53_exon_8 TP53 ENSG00000141510 ENST00000269305
    chr17 7674177 7674293 TP53_exon_7 TP53 ENSG00000141510 ENST00000269305
    chr17 7674855 7674974 TP53_exon_6 TP53 ENSG00000141510 ENST00000269305
    chr17 7675049 7675239 TP53_exon_5 TP53 ENSG00000141510 ENST00000269305
    chr17 7675990 7676275 TP53_exon_4 TP53 ENSG00000141510 ENST00000269305
    chr17 7676378 7676406 TP53_exon_3 TP53 ENSG00000141510 ENST00000269305
    chr17 7676517 7676597 TP53_exon_2 TP53 ENSG00000141510 ENST00000269305
    chr19 12943710 12943913 CALR_exon_9 + CALR ENSG00000179218 ENST00000316448
    chr19 33301337 33302417 CEBPA_exon_1 CEBPA ENSG00000245848 ENST00000498907
    chr2 197392302 197392464 SF3B1_exon_25 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197392968 197393191 SF3B1_exon_24 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197396052 197396331 SF3B1_exon_23 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197397981 197398119 SF3B1_exon_22 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197398457 197398584 SF3B1_exon_21 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197400051 197400169 SF3B1_exon_20 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197400248 197400437 SF3B1_exon_19 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197400711 197400939 SF3B1_exon_18 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197401396 197401528 SF3B1_exon_17 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197401738 197401891 SF3B1_exon_16 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197401981 197402133 SF3B1_exon_15 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197402552 197402829 SF3B1_exon_14 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197402945 197403038 SF3B1_exon_13 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197403581 197403767 SF3B1_exon_12 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197405072 197405180 SF3B1_exon_11 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197405271 197405475 SF3B1_exon_10 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197407994 197408122 SF3B1_exon_9 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197408365 197408584 SF3B1_exon_8 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197409766 197410010 SF3B1_exon_7 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197416737 197416914 SF3B1_exon_6 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197418505 197418591 SF3B1_exon_5 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197420424 197420545 SF3B1_exon_4 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197421025 197421136 SF3B1_exon_3 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197423804 197423977 SF3B1_exon_2 SF3B1 ENSG00000115524 ENST00000335508
    chr2 197434968 197435002 SF3B1_exon_1 SF3B1 ENSG00000115524 ENST00000335508
    chr2 208248366 208248663 IDH1_exon_4 IDH1 ENSG00000138413 ENST00000415913
    chr2 25234278 25234423 DNMT3A_exon_23 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25235703 25235828 DNMT3A_exon_22 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25236932 25237008 DNMT3A_exon_21 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25239126 25239218 DNMT3A_exon_20 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25240298 25240453 DNMT3A_exon_19 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25240636 25240733 DNMT3A_exon_18 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25241558 25241710 DNMT3A_exon_17 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25243894 25243985 DNMT3A_exon_16 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25244151 25244341 DNMT3A_exon_15 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25244536 25244655 DNMT3A_exon_14 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25245249 25245335 DNMT3A_exon_13 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25246016 25246067 DNMT3A_exon_12 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25246156 25246312 DNMT3A_exon_11 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25246616 25246779 DNMT3A_exon_10 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25247047 25247161 DNMT3A_exon_9 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25247587 25247752 DNMT3A_exon_8 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25248033 25248255 DNMT3A_exon_7 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25274937 25275090 DNMT3A_exon_6 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25275496 25275546 DNMT3A_exon_5 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25282437 25282714 DNMT3A_exon_4 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25300135 25300246 DNMT3A_exon_3 DNMT3A ENSG00000119772 ENST00000264709
    chr2 25313909 25313987 DNMT3A_exon_2 DNMT3A ENSG00000119772 ENST00000264709
    chr20 32358772 32358835 ASXL1_exon_1 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32366380 32366469 ASXL1_exon_2 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32367723 32367732 ASXL1_exon_3 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32369011 32369126 ASXL1_exon_4 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32428124 32428251 ASXL1_exon_5 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32428321 32428425 ASXL1_exon_6 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32429334 32429434 ASXL1_exon_7 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32429897 32430056 ASXL1_exon_8 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32431317 32431487 ASXL1_exon_9 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32431579 32431682 ASXL1_exon_10 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32432876 32432988 ASXL1_exon_11 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32433280 32433920 ASXL1_exon_12 + ASXL1 ENSG00000171456 ENST00000375687
    chr20 32434428 32437338 ASXL1_exon_13 + ASXL1 ENSG00000171456 ENST00000375687
    chr21 34792134 34792613 RUNX1_exon_8 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34799297 34799465 RUNX1_exon_7 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34834406 34834604 RUNX1_exon_6 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34859470 34859581 RUNX1_exon_5 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34880553 34880716 RUNX1_exon_4 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34886839 34887099 RUNX1_exon_3 RUNX1 ENSG00000159216 ENST00000300305
    chr21 34892921 34892966 RUNX1_exon_2 RUNX1 ENSG00000159216 ENST00000300305
    chr21 35048838 35048902 RUNX1_exon_1 RUNX1 ENSG00000159216 ENST00000300305
    chr21 43094652 43094791 U2AF1_exon_6 U2AF1 ENSG00000160201 ENST00000291552
    chr21 43104312 43104405 U2AF1_exon_2 U2AF1 ENSG00000160201 ENST00000291552
    chr3 128481018 128481321 GATA2_exon_6 GATA2 ENSG00000179348 ENST00000341105
    chr3 128481815 128481947 GATA2_exon_5 GATA2 ENSG00000179348 ENST00000341105
    chr3 128483856 128484008 GATA2_exon_4 GATA2 ENSG00000179348 ENST00000341105
    chr3 128485723 128486371 GATA2_exon_3 GATA2 ENSG00000179348 ENST00000341105
    chr3 128486799 128487034 GATA2_exon_2 GATA2 ENSG00000179348 ENST00000341105
    chr4 105233939 105237354 TET2_exon_3 + TET2 ENSG00000168769 ENST00000540549
    chr4 105241335 105241432 TET2_exon_4 + TET2 ENSG00000168769 ENST00000540549
    chr4 105242830 105242930 TET2_exon_5 + TET2 ENSG00000168769 ENST00000540549
    chr4 105243566 105243781 TET2_exon_6 + TET2 ENSG00000168769 ENST00000540549
    chr4 105259615 105259772 TET2_exon_7 + TET2 ENSG00000168769 ENST00000540549
    chr4 105261755 105261851 TET2_exon_8 + TET2 ENSG00000168769 ENST00000540549
    chr4 105269606 105269750 TET2_exon_9 + TET2 ENSG00000168769 ENST00000540549
    chr4 105272560 105272921 TET2_exon_10 + TET2 ENSG00000168769 ENST00000540549
    chr4 105275044 105276519 TET2_exon_11 + TET2 ENSG00000168769 ENST00000540549
    chr4 54695509 54695784 KIT_exon_2 + KIT ENSG00000157404 ENST00000288135
    chr4 54723581 54723701 KIT_exon_8 + KIT ENSG00000157404 ENST00000288135
    chr4 54725854 54726053 KIT_exon_9 + KIT ENSG00000157404 ENST00000288135
    chr4 54727215 54727327 KIT_exon_10 + KIT ENSG00000157404 ENST00000288135
    chr4 54727413 54727545 KIT_exon_11 + KIT ENSG00000157404 ENST00000288135
    chr4 54728008 54728124 KIT_exon_13 + KIT ENSG00000157404 ENST00000288135
    chr4 54733067 54733195 KIT_exon_17 + KIT ENSG00000157404 ENST00000288135
    chr5 171410524 171410565 NPM1_exon_11 + NPM1 ENSG00000181163 ENST00000296930
    chr7 101816027 101816096 CUX1_exon_1 + CUX1 ENSG00000257923 ENST00000360264
    chr7 101916111 101916228 CUX1_exon_2 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102028094 102028148 CUX1_exon_3 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102070335 102070420 CUX1_exon_4 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102097360 102097504 CUX1_exon_5 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102104332 102104462 CUX1_exon_6 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102111694 102111777 CUX1_exon_7 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102115203 102115276 CUX1_exon_8 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102158556 102158611 CUX1_exon_9 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102170442 102170553 CUX1_exon_10 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102178465 102178660 CUX1_exon_11 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102189809 102189874 CUX1_exon_12 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102193838 102193893 CUX1_exon_13 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102195503 102195606 CUX1_exon_14 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102196630 102197308 CUX1_exon_15 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102198798 102198870 CUX1_exon_16 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102200067 102200175 CUX1_exon_17 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102201356 102202207 CUX1_exon_18 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102204387 102204559 CUX1_exon_19 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102205110 102205173 CUX1_exon_20 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102227363 102227672 CUX1_exon_21 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102234048 102234243 CUX1_exon_22 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102239316 102239587 CUX1_exon_23 + CUX1 ENSG00000257923 ENST00000360264
    chr7 102248408 102249042 CUX1_exon_24 + CUX1 ENSG00000257923 ENST00000360264
    chr7 140753272 140753396 BRAF_exon_15 BRAF ENSG00000157764 ENST00000288602
    chr7 148807645 148807709 EZH2_exon_20 EZH2 ENSG00000106462 ENST00000320356
    chr7 148809067 148809158 EZH2_exon_19 EZH2 ENSG00000106462 ENST00000320356
    chr7 148809306 148809393 EZH2_exon_18 EZH2 ENSG00000106462 ENST00000320356
    chr7 148810329 148810417 EZH2_exon_17 EZH2 ENSG00000106462 ENST00000320356
    chr7 148811621 148811723 EZH2_exon_16 EZH2 ENSG00000106462 ENST00000320356
    chr7 148813955 148814140 EZH2_exon_15 EZH2 ENSG00000106462 ENST00000320356
    chr7 148814910 148815042 EZH2_exon_14 EZH2 ENSG00000106462 ENST00000320356
    chr7 148815502 148815549 EZH2_exon_13 EZH2 ENSG00000106462 ENST00000320356
    chr7 148816680 148816781 EZH2_exon_12 EZH2 ENSG00000106462 ENST00000320356
    chr7 148817218 148817394 EZH2_exon_11 EZH2 ENSG00000106462 ENST00000320356
    chr7 148817873 148818120 EZH2_exon_10 EZH2 ENSG00000106462 ENST00000320356
    chr7 148819592 148819690 EZH2_exon_9 EZH2 ENSG00000106462 ENST00000320356
    chr7 148826450 148826635 EZH2_exon_8 EZH2 ENSG00000106462 ENST00000320356
    chr7 148827160 148827269 EZH2_exon_7 EZH2 ENSG00000106462 ENST00000320356
    chr7 148828736 148828883 EZH2_exon_6 EZH2 ENSG00000106462 ENST00000320356
    chr7 148829724 148829851 EZH2_exon_5 EZH2 ENSG00000106462 ENST00000320356
    chr7 148832630 148832753 EZH2_exon_4 EZH2 ENSG00000106462 ENST00000320356
    chr7 148846466 148846601 EZH2_exon_3 EZH2 ENSG00000106462 ENST00000320356
    chr7 148847178 148847301 EZH2_exon_2 EZH2 ENSG00000106462 ENST00000320356
    chr8 116847499 116847694 RAD21_exon_14 RAD21 ENSG00000164754 ENST00000297338
    chr8 116848942 116849032 RAD21_exon_13 RAD21 ENSG00000164754 ENST00000297338
    chr8 116850614 116850770 RAD21_exon_12 RAD21 ENSG00000164754 ENST00000297338
    chr8 116851944 116852099 RAD21_exon_11 RAD21 ENSG00000164754 ENST00000297338
    chr8 116852545 116852711 RAD21_exon_10 RAD21 ENSG00000164754 ENST00000297338
    chr8 116854241 116854471 RAD21_exon_9 RAD21 ENSG00000164754 ENST00000297338
    chr8 116856162 116856291 RAD21_exon_8 RAD21 ENSG00000164754 ENST00000297338
    chr8 116856642 116856774 RAD21_exon_7 RAD21 ENSG00000164754 ENST00000297338
    chr8 116857263 116857476 RAD21_exon_6 RAD21 ENSG00000164754 ENST00000297338
    chr8 116858348 116858461 RAD21_exon_5 RAD21 ENSG00000164754 ENST00000297338
    chr8 116861837 116861943 RAD21_exon_4 RAD21 ENSG00000164754 ENST00000297338
    chr8 116863126 116863262 RAD21_exon_3 RAD21 ENSG00000164754 ENST00000297338
    chr8 116866582 116866732 RAD21_exon_2 RAD21 ENSG00000164754 ENST00000297338
    chr9 5069922 5070055 JAK2_exon_12 + JAK2 ENSG00000096968 ENST00000381652
    chr9 5073695 5073788 JAK2_exon_14 + JAK2 ENSG00000096968 ENST00000381652
    chrX 124022624 124022674 STAG2_exon_3 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124025836 124025921 STAG2_exon_4 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124030957 124031128 STAG2_exon_5 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124037523 124037626 STAG2_exon_6 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124042565 124042648 STAG2_exon_7 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124045160 124045371 STAG2_exon_8 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124047350 124047508 STAG2_exon_9 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124049001 124049081 STAG2_exon_10 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124050182 124050312 STAG2_exon_11 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124051117 124051222 STAG2_exon_12 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124051311 124051397 STAG2_exon_13 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124056124 124056238 STAG2_exon_14 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124057862 124057980 STAG2_exon_15 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124061220 124061344 STAG2_exon_16 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124061767 124061877 STAG2_exon_17 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124062898 124062997 STAG2_exon_18 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124063112 124063208 STAG2_exon_19 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124063844 124064054 STAG2_exon_20 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124065872 124065949 STAG2_exon_21 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124066171 124066265 STAG2_exon_22 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124066352 124066439 STAG2_exon_23 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124068560 124068659 STAG2_exon_24 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124071145 124071326 STAG2_exon_25 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124076328 124076474 STAG2_exon_26 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124077953 124078061 STAG2_exon_27 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124081376 124081531 STAG2_exon_28 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124083417 124083552 STAG2_exon_29 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124086543 124086773 STAG2_exon_30 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124090571 124090767 STAG2_exon_31 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124090850 124090967 STAG2_exon_32 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124094014 124094147 STAG2_exon_33 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124095368 124095452 STAG2_exon_34 + STAG2 ENSG00000101972 ENST00000218089
    chrX 124100570 124100597 STAG2_exon_35 + STAG2 ENSG00000101972 ENST00000218089
    chrX 130005228 130005320 BCORL1_exon_1 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130012574 130012671 BCORL1_exon_2 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130012946 130016216 BCORL1_exon_3 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130020981 130021153 BCORL1_exon_4 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130022893 130022980 BCORL1_exon_5 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130024986 130025382 BCORL1_exon_6 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130028631 130028864 BCORL1_exon_7 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130037363 130037536 BCORL1_exon_8 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130039133 130039285 BCORL1_exon_9 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130050713 130050797 BCORL1_exon_10 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130051856 130052019 BCORL1_exon_11 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 130055850 130056136 BCORL1_exon_12 + BCORL1 ENSG00000085185 ENST00000540052
    chrX 134377614 134377758 PHF6_exon_2 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134378001 134378109 PHF6_exon_3 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134393497 134393637 PHF6_exon_4 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134393905 134393955 PHF6_exon_5 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134413487 134413660 PHF6_exon_6 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134413819 134413969 PHF6_exon_7 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134415012 134415123 PHF6_exon_8 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134417165 134417305 PHF6_exon_9 + PHF6 ENSG00000156531 ENST00000332070
    chrX 134425197 134425330 PHF6_exon_10 + PHF6 ENSG00000156531 ENST00000332070
    chrX 15321505 15321775 PIGA_exon_6 PIGA ENSG00000165195 ENST00000333590
    chrX 15324661 15324874 PIGA_exon_5 PIGA ENSG00000165195 ENST00000333590
    chrX 15325016 15325155 PIGA_exon_4 PIGA ENSG00000165195 ENST00000333590
    chrX 15325910 15326049 PIGA_exon_3 PIGA ENSG00000165195 ENST00000333590
    chrX 15331212 15331933 PIGA_exon_2 PIGA ENSG00000165195 ENST00000333590
    chrX 15790492 15790539 ZRSR2_exon_1 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15790930 15791016 ZRSR2_exon_2 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15799868 15799956 ZRSR2_exon_3 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15803684 15803799 ZRSR2_exon_4 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15804107 15804200 ZRSR2_exon_5 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15808229 15808274 ZRSR2_exon_6 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15809196 15809321 ZRSR2_exon_7 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15815673 15815893 ZRSR2_exon_8 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15818583 15818645 ZRSR2_exon_9 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15820203 15820319 ZRSR2_exon_10 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 15822727 15823242 ZRSR2_exon_11 + ZRSR2 ENSG00000169249 ENST00000307771
    chrX 40052108 40052403 BCOR_exon_15 BCOR ENSG00000183337 ENST00000378444
    chrX 40053882 40054045 BCOR_exon_14 BCOR ENSG00000183337 ENST00000378444
    chrX 40054252 40054336 BCOR_exon_13 BCOR ENSG00000183337 ENST00000378444
    chrX 40055364 40055516 BCOR_exon_12 BCOR ENSG00000183337 ENST00000378444
    chrX 40057151 40057324 BCOR_exon_11 BCOR ENSG00000183337 ENST00000378444
    chrX 40062135 40062396 BCOR_exon_10 BCOR ENSG00000183337 ENST00000378444
    chrX 40062742 40063074 BCOR_exon_9 BCOR ENSG00000183337 ENST00000378444
    chrX 40063604 40063955 BCOR_exon_8 BCOR ENSG00000183337 ENST00000378444
    chrX 40064332 40064602 BCOR_exon_7 BCOR ENSG00000183337 ENST00000378444
    chrX 40070969 40071162 BCOR_exon_6 BCOR ENSG00000183337 ENST00000378444
    chrX 40071633 40071693 BCOR_exon_5 BCOR ENSG00000183337 ENST00000378444
    chrX 40072345 40075183 BCOR_exon_4 BCOR ENSG00000183337 ENST00000378444
    chrX 40076450 40076535 BCOR_exon_3 BCOR ENSG00000183337 ENST00000378444
    chrX 40077840 40077932 BCOR_exon_2 BCOR ENSG00000183337 ENST00000378444
    chrX 53380102 53380189 SMC1A_exon_25 SMC1A ENSG00000072501 ENST00000322213
    chrX 53380616 53380733 SMC1A_exon_24 SMC1A ENSG00000072501 ENST00000322213
    chrX 53381014 53381090 SMC1A_exon_23 SMC1A ENSG00000072501 ENST00000322213
    chrX 53382228 53382386 SMC1A_exon_22 SMC1A ENSG00000072501 ENST00000322213
    chrX 53382502 53382663 SMC1A_exon_21 SMC1A ENSG00000072501 ENST00000322213
    chrX 53383093 53383256 SMC1A_exon_20 SMC1A ENSG00000072501 ENST00000322213
    chrX 53394774 53394891 SMC1A_exon_19 SMC1A ENSG00000072501 ENST00000322213
    chrX 53396223 53396383 SMC1A_exon_18 SMC1A ENSG00000072501 ENST00000322213
    chrX 53396468 53396620 SMC1A_exon_17 SMC1A ENSG00000072501 ENST00000322213
    chrX 53399585 53399733 SMC1A_exon_16 SMC1A ENSG00000072501 ENST00000322213
    chrX 53403562 53403675 SMC1A_exon_15 SMC1A ENSG00000072501 ENST00000322213
    chrX 53403773 53403896 SMC1A_exon_14 SMC1A ENSG00000072501 ENST00000322213
    chrX 53405008 53405152 SMC1A_exon_13 SMC1A ENSG00000072501 ENST00000322213
    chrX 53405241 53405394 SMC1A_exon_12 SMC1A ENSG00000072501 ENST00000322213
    chrX 53405489 53405675 SMC1A_exon_11 SMC1A ENSG00000072501 ENST00000322213
    chrX 53405767 53405959 SMC1A_exon_10 SMC1A ENSG00000072501 ENST00000322213
    chrX 53409058 53409272 SMC1A_exon_9 SMC1A ENSG00000072501 ENST00000322213
    chrX 53409417 53409506 SMC1A_exon_8 SMC1A ENSG00000072501 ENST00000322213
    chrX 53411757 53411904 SMC1A_exon_7 SMC1A ENSG00000072501 ENST00000322213
    chrX 53411991 53412256 SMC1A_exon_6 SMC1A ENSG00000072501 ENST00000322213
    chrX 53412896 53413141 SMC1A_exon_5 SMC1A ENSG00000072501 ENST00000322213
    chrX 53413228 53413438 SMC1A_exon_4 SMC1A ENSG00000072501 ENST00000322213
    chrX 53414754 53414873 SMC1A_exon_3 SMC1A ENSG00000072501 ENST00000322213
    chrX 53414977 53415172 SMC1A_exon_2 SMC1A ENSG00000072501 ENST00000322213
    chrX 53422488 53422603 SMC1A_exon_1 SMC1A ENSG00000072501 ENST00000322213
  • Confirmatory Studies
  • We used FISH, PCR, and chromosomal microarray analyses, with or without existing RNA-sequencing data, to confirm findings on whole-genome sequencing that had not been detected by cytogenetic analysis. We used standard protocols to perform chromosomal microarray analysis in the Washington University Cytogenetics Core. In the PCR-confirmation analyses, we used primers designed to detect structural variant breakpoints. The methods that were used in RNA sequencing for structural variants in selected samples have been reported previously.
  • WGS results were compared to conventional cytogenetics and FISH to determine the sensitivity and positive predictive value for detecting recurrent SVs and CNAs. These comparisons used the following approaches:
      • SVs: Cases with successful cytogenetics (at least 3 metaphases analyzed, N=235) were used to evaluate SV performance. SVs identified by WGS were manually compared to ISCN karyotypes obtained from clinical testing to identify true positives. Breakpoints were required to occur within 1 chromosome band. New SVs that were not reported by conventional cytogenetics were subject to confirmation using either FISH, PCR for SV breakpoints, or analysis of existing RNA-seq data for fusion transcripts (see below).
      • CNAs: CNAs from WGS were compared to ISCN karyotypes using 143 cases with conclusive cytogenetic results (i.e., 20 metaphases) and no ambiguous findings, such as composite karyotypes, marker chromosomes, or additional unidentifiable material, as these preclude definitive comparisons. ISCN cytogenetic karyotypes were transformed into a matrix of gains and losses for each chromosome band using published software, which were then converted with a custom PERL script to BEDPE format using band coordinates based on the GRCh38 human reference. The bedtools program was then used to compare CNAs between WGS and cytogenetics using at least 1 bp of overlap to identify concordant events. New CNAs were subject to confirmation using either FISH or chromosomal microarrays (CMA).
  • Every effort was made to confirm all novel findings, although priority was given to findings in the prospective cohort and for risk-defining events. Specific confirmation procedures are described below.
  • FISH
  • WGS findings not present in the karyotype or confirmed by diagnostic FISH results were confirmed using FISH studies where possible. FISH was the primary means of confirmation for new SVs and CNAs when appropriate probes were available and clinical specimens were available for testing. All FISH studies were performed using validated probes and standard clinical procedures using 200 cells and were reviewed by board certified cytogeneticists. The presence of an abnormal result in the specified study was considered as support for the genomic event identified by WGS. For example, we considered an abnormal result for the KMT2A dual color/dual fusion FISH assay as confirmation of an SV involving KTM2A in the WGS data.
  • PCR
  • Selected SVs that could not be confirmed via FISH because of insufficient or inadequate samples were confirmed via PCR from DNA using primers spanning the SV breakends identified by Manta when FISH studies could not be performed due to limited material and/or lack of appropriate FISH probes. PCR primers were designed from breakpoint-spanning sequence contigs generated from Manta and were used in standard PCR reactions with human genomic DNA. Amplified fragments were excised, purified, sequenced with Sanger sequencing, and analyzed with Blat to verify localization to the breakpoint region.
  • CMA
  • CNAs were confirmed via chromosomal microarray (CMA) for cases with available DNA but insufficient material or probe for FISH assays. CMAs were performed per standard methods using the CytoScan HD platform (ThermoFisher) with subsequent analysis in Chromosome Analysis Suite (ThermoFisher). Data were reviewed and interpreted by a board-certified cytogeneticist and molecular geneticist.
  • RNA-Seq
  • SVs in two cases with KMT2A rearrangements were confirmed using existing RNA-seq data that was published as part of the TCGA AML study (see Supplemental Table 1 in ref 18, which can be accessed here: https://api.gdc.cancer.gov/data/b9196563-a05d-40b8-80dc-640ec712eb06; samples 380949 and 410324). We note that clinical FISH using a KMT2A breakapart probe for these cases was also abnormal, and the identification of a fusion transcript via RNA-seq provided the definitive confirmation of the translocation partner.
  • Risk Stratification
  • Conventional
  • T provide a basis of comparison for risk stratification results obtained using the disclosed WGS method, cytogenetics, FISH, and molecular results were used to assign patients to established genomic risk categories, which used the 2017 ELN guidelines for AML patients12 and the cytogenetic component of the IPSS-R scoring system for MDS patients, both without modification. Cytogenetic abnormalities were required to meet the abovementioned criteria to be considered clonal. For AML patients, risk group assignment was performed using cytogenetic results, FLT3 ITD mutation allele ratio from PCR (or presence/absence if the allelic ratio was not available), and the mutation status for CEBPA, NPM1, TP53, RUNX1, and ASXL1 from either clinical tumor/normal exome sequencing (N=12) or gene panel sequencing (using Myeloseq, N=71, or a commercial assay, N=1). Sequencing assays were not performed for 6 retrospective patients who were either assigned to a risk group using only NPM1 and FLT3 ITD mutation status (N=3), or they were assigned to intermediate risk (N=3). Patients with a normal karyotype and <20 metaphases were not assigned to a risk group with unless there was an unequivocal result from either FISH or targeted sequencing (e.g., a positive PML-RARA or del(5q) by FISH, or a TP53 mutation by targeted sequencing). IPSS-R risk groups do not involve gene mutations and are therefore performed using cytogenetics alone.
  • WGS
  • WGS results were used to assign patients to risk groups using the identical guidelines as above for both AML and MDS patients. For AML patients, risk assignment used CNAs, recurrent SVs, and gene mutations. FLT3 ITD mutation results from PCR were used instead of the WGS result (even though ITD alleles can be detected) because the PCR assay is an FDA-cleared companion diagnostic for the FLT3 targeted therapy midostaurin. For both AML and MDS patients, the clinically important classifications of normal karyotype and complex karyotype used only CNAs and recurrent SVs and not SVs reported as secondary findings. A normal karyotype was designated if no variants in either category were identified, and a complex karyotype was designated if at least 3 chromosomal abnormalities were identified, including recurrent SVs (not WHO category-defining events) or CNAs greater than 5 Mbp that were identified by copy number analysis and that involved separate chromosome arms. All but 3 of the patients with a complex karyotype could be assigned to this category based on CNAs alone, which indicates that copy number gains and losses are defining features of this phenotype.
  • Statistics
  • Statistical Analysis
  • In the time-to-event survival analysis involving study patients with AML, we used death as the end point for the Kaplan-Meier analysis or Cox proportional hazards regression to test for equal survival across genetic risk groups. Censoring of patients in these analyses was random and occurred because of limited follow-up time. Survival analyses of patients with defined cytogenetic risk (N=71 nontransplanted patients; N=101 total patients) was pre-planned using patients within our cohort (i.e., they were not selected specifically for outcome analysis) and was performed by Kaplan-Meier analyses using the log-rank test for equal survival across the groups. Cox proportional-hazards regression was used to calculate hazard ratios and test for equal survival between the adverse risk group and either intermediate, favorable, or a combined intermediate/favorable ‘not adverse’ risk group. All log-rank tests performed in the paper were adjusted for multiple comparisons using the method of Benjamini and Hochberg (1995). Cox regression was adjusted for age (binned by decade), which was significantly associated with overall survival in the 71 non-transplanted patients with defined risk stratified by conventional risk groups (HR: 1.46, 95% CI 1.05-2.05) but not WGS-based risk groups (HR: 1.29, 95% CI 0.92-1.81). The log of the white blood cell count was also used as a covariate with ELN risk, but was not significant in any analysis (P>0.05 in all analyses) and therefore was not included in the model. The proportional hazards assumption was found to be tenable for all Cox models.
  • The same approaches were used for AML patients with undefined cytogenetic risk (N=27 nontransplanted patients; N=38 total patients). Prior to this pre-planned analysis, we performed a power calculation to estimate the sample size necessary to observe a difference in survival among ELN risk groups in this cohort. This used the Power and Sample Size task in SAS/Studio software along with the observed survival in the defined cytogenetic risk cohort above (N=71), which was largely consistent with published data on a mixture of older (60 and over) and younger (less than 60) patients. The power calculation used a median survival of 3600 days of survival for the favorable group and 346 for the adverse group, with a minimum follow-up interval of 279 days and a total number of days (accrual+follow-up) of 750 days. This demonstrated 80% power to detect a survival difference between favorable and adverse risk at a sample size of 12 (per group) using an alpha of 0.05. Additional exploratory analyses were performed but not presented, including log-rank tests for differences in survival among all three risk groups (rather than not adverse vs. adverse) and unadjusted Cox regression tests, which yielded similar results to those shown here. Survival statistics were obtained using SAS for Windows, Version 9.4. The survminer package in R was used for visualization.
  • Results Streamlined Approach to Whole-Genome Sequencing
  • We developed a streamlined approach to whole-genome sequencing (ChromoSeq) that was designed to provide comprehensive genomic profiling of clinically relevant mutations in samples obtained from patients with AML or MDS, while minimizing the turnaround time and technical complexity (FIG. 5). In this approach, we used scalable methods of sample preparation that can be performed by a single technician in less than 8 hours with commercially available reagents, followed by standard high-throughput sequencing. Automated tumor-only variant analysis detected mutations in selected genes, copy-number alterations of more than 5 Mbp, and recurrent structural variants (Tables S1 and S2, above). We then summarized these findings in a concise clinical report (FIG. 9).
  • We performed a head-to-head comparison of this approach with conventional cytogenetic analysis and targeted sequencing using 235 samples obtained from patients with a known or suspected hematologic cancer who had undergone successful cytogenetic analysis. This sequencing analysis yielded a mean genome coverage of 50×; a mean of 5.1 clinically relevant mutations (range, 0 to 20) were detected per patient across all variant types (FIGS. 10 and 11). The sensitivity of whole-genome sequencing for recurrent translocations that had been reported on cytogenetic analysis was 100% (40 of 40 samples) (FIG. 6A).
  • Whole-genome sequencing identified cytogenetically cryptic structural variants in 13 patients, including complex or cryptic chromosomal translocations involving the inv(16)(p13.1q22) fusion gene CBFB-MYH11 in 2 patients, the t(7;21)(p22;q22) fusion gene USP42-RUNX1 in 1 patient, and 10 rearrangements involving KMT2A, all of which were verified with the use of orthogonal methods (FIG. 6B and FIG. 12, Whole-genome sequencing detected 100% (91 of 91) of the clonal copy-number alterations that had been detected on cytogenetic analysis among the 143 patients in whom conclusive and unambiguous results had been identified by karyotyping (FIG. 6A). In addition, sequencing identified 21 new copy-number alterations in 14 of these patients, 12 of which were confirmed by other methods (FIG. 6C). The remaining 9 new copy-number alterations showed altered coverage patterns on whole-genome sequencing but could not be confirmed by orthogonal methods because of their small size, low abundance, or both (FIGS. 6C, 14A, 14B, and 14C). Whole-genome sequencing also provided definitive identification of copy-number alterations in an additional 13 patients with ambiguous or inconclusive results by cytogenetic analysis (Table S5). When we combined these results with the findings in 14 patients who had conclusive results by cytogenetic analysis and newly identified copy-number alterations, plus the findings in 13 patients who were identified as having new structural variants (see Table S4), we determined that 40 of 235 patients (17.0%) had results that had not been detected by conventional cytogenetic analysis.
  • TABLE S5
    New CNAs Identified by WGS
    Chrom Start End Size Bands Type Diagnosis WGS.CNAs WGS.Recurrent.SVs
    chr16 61500000 90000000 28500000 q21qter DEL AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr17 500000 10500000 1.00E+07 pterp13.1 DEL AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr18 500000 80000000 79500000 pterqter DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr22 17500000 36500000 1.90E+07 q11.21q12.3 DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr3 1000000 87500000 86500000 pterp11.2 DEL AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr5 1000000 52000000 5.10E+07 pterq11.2 DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr5 52000000 181000000 1.29E+08 q11.2qter DEL AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr7 500000 159000000 158500000 pterqter DEL AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr8 500000 145000000 144500000 pterqter DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr9 500000 138000000 137500000 pterqter DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr15 23500000 101500000 7.80E+07 q11.2qter DUP AML del(3)(p11.2pter)[61.8%], +5[76.7%], 0
    del(5)(q11.2qter)[60.6%], −7[57.2%], +8[57.6%], +9[52.3%],
    gain(15)(q11.2qter)[43.2%], del(16)(q21qter)[58.5%],
    del(17)(p13.1pter)[53.0%], +18[43.5%], gain(22)(q11.21q12.3)[64.0%]
    chr13 67000000 113500000 46500000 q21.32qter DUP AML del(7)(q22.1qter)[60.0%], del(10)(q22.2q22.3)[58.6%], gain(13)(q21.32qter)[56.6%] 0
    chr7 101500000 159000000 57500000 q22.1qter DEL AML del(7)(q22.1qter)[60.0%], del(10)(q22.2q22.3)[58.6%], gain(13)(q21.32qter)[56.6%] 0
    chr10 75000000 79500000 4500000 q22.2q22.3 DEL AML del(7)(q22.1qter)[60.0%], del(10)(q22.2q22.3)[58.6%], gain(13)(q21.32qter)[56.6%] 0
    chr13 60500000 62500000 2.00E+06 q21.2q21.31 DEL AML del(13)(q21.2q21.31)[96.6%] 0
    chr11 117000000 135000000 1.80E+07 q23.3qter DUP AML gain(11)(q23.3qter)[15.0%] inv(16)(q22.1p13.11)[36.5%]
    chr1 3000000 248000000 2.45E+08 p36.32qter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr12 500000 133000000 132500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr13 20000000 113500000 93500000 q12.11qter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr15 23500000 101500000 7.80E+07 q11.2qter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr16 1500000 90000000 88500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr17 500000 83000000 82500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr18 500000 80000000 79500000 pterqter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr19 1500000 58500000 5.70E+07 pterqter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr2 500000 242000000 241500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr20 500000 64000000 63500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr21 14000000 46500000 32500000 q11.2qter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr3 1000000 197500000 196500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr4 500000 189500000 1.89E+08 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr5 104000000 181000000 7.70E+07 q21.2qter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr6 500000 170500000 1.70E+08 pterqter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr7 500000 159000000 158500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr8 500000 145000000 144500000 pterqter DUP ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr9 500000 138000000 137500000 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chrX 3000000 154000000 1.51E+08 pterqter DEL ALL +1[44.6%], −2[21.4%], −3[21.6%], −4[22.0%], 0
    del(5)(q21.2qter)[20.7%], +6[42.6%], −7[21.7%], +8[42.3%], −9[20.8%], −12[21.3%],
    del(13)(q12.11qter)[21.5%],
    del(15)(q11.2qter)[20.7%], −16[19.7%], −17[19.2%], +18[42.1%], +19[47.4%], −20[19.1%],
    gain(21)(q11.2qter)[43.2%], −X[2
    chr11 72500000 135000000 62500000 q13.4qter DUP AML del(2)(q36.3qter)[89.1%], gain(11)(q13.4qter)[90.2%] 0
    chr2 227500000 242000000 14500000 q36.3qter DEL AML del(2)(q36.3qter)[89.1%], gain(11)(q13.4qter)[90.2%] 0
    chr8 500000 39500000 3.90E+07 pterp11.22 DUP MDS gain(8)(p11.22pter)[160.7%], −8[82.0%], gain(8)(q12.3qter)[155.8%] 0
    chr8 39500000 64000000 24500000 p11.22q12.3 DEL MDS gain(8)(p11.22pter)[160.7%], −8[82.0%], gain(8)(q12.3qter)[155.8%] 0
    chr8 64000000 145000000 8.10E+07 q12.3qter DUP MDS gain(8)(p11.22pter)[160.7%], −8[82.0%], gain(8)(q12.3qter)[155.8%] 0
    chr18 500000 14000000 13500000 pterp11.21 DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr19 1500000 20000000 18500000 pterp12 DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr20 31500000 64000000 32500000 q11.21qter DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr7 38500000 48000000 9500000 p14.1p12.3 DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr7 48000000 159000000 1.11E+08 p12.3qter DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr13 20000000 113500000 93500000 q12.11qter DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr16 74500000 90000000 15500000 q23.1qter DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr9 500000 32500000 3.20E+07 pterp21.1 DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%1
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%1
    chr2 118000000 242000000 1.24E+08 q14.1qter DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr9 99500000 138000000 38500000 q22.33qter DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr10 500000 37000000 36500000 pterp11.21 DEL AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chr10 43000000 133500000 90500000 q11.21qter DUP AML gain(2)(q14.1qter)[23.6%], gain(7)(p12.3p14.1)[23.6%], −7[21.5%], t(15; 17)(q24.1; q21.2)[3.3%],
    del(9)(p21.1pter)[17.4%], gain(9)(q22.33qter)[21.4%], del(10)(p11.21pter)[23.8%], t(15; 17)(q24.1; q21.2)[4.1%]
    gain(10)(q11.21qter)[22.1%], del(13)(q12.11qter)[20.7%], del(16)(q23.1qter)[21.7%],
    del(18)(p11.21pter)[15.2%], gain(19)(p12pter)[21.7%], gain(20)(q11.21qter)[35.8%]
    chrY 7000000 21000000 1.40E+07 p11.2q11.223 DEL MDS del(4)(q21.1q25)[53.3%], −Y[27.1%] 0
    chr4 76000000 107000000 3.10E+07 q21.1q25 DEL MDS del(4)(q21.1q25)[53.3%], −Y[27.1%] 0
    chrX 3000000 9500000 6500000 pterp22.31 DUP AML del(5)(q31.2q31.2)[10.1%] 0
    chr5 137577914 139513006 1935093 q31.2q31.2 DEL AML del(5)(q31.2q31.2)[10.1%] 0
    chr19 2000000 58500000 56500000 pterqter DUP AML del(5)(q31.2q31.2)[10.1%] 0
    chr5 1000000 30000000 2.90E+07 pterp13.3 DUP AML del(5)(q31.2q31.2)[10.1%] 0
    chr7 92000000 159000000 6.70E+07 q21.2qter DEL MDS del(5)(q11.2qter)[63.2%], del(7)(q21.2qter)[63.2%], +8[43.1%1 0
    chr8 500000 145000000 144500000 pterqter DUP MDS del(5)(q11.2qter)[63.2%], del(7)(q21.2qter)[63.2%], +8[43.1%] 0
    chr5 57500000 181000000 123500000 q11.2qter DEL MDS del(5)(q11.2qter)[63.2%], del(7)(q21.2qter)[63.2%], +8[43.1%] 0
    chr21 41000000 46500000 5500000 q22.2qter DUP AML +4[7.7%], gain(21)(q22.2qter)[10.7%] 0
    chr4 500000 189500000 1.89E+08 pterqter DUP AML +4[7.7%], gain(21)(q22.2qter)[10.7%] 0
    chr13 31000000 104000000 7.30E+07 q12.3q33.1 DEL ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chr8 500000 145000000 144500000 pterqter DUP ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chrX 3000000 154000000 1.51E+08 pterqter DUP ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chr7 500000 54500000 5.40E+07 pterp11.2 DEL ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chr14 20000000 105500000 85500000 q11.2qter DUP ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chr3 1000000 197500000 196500000 pterqter DUP ALL +3[26.4%], del(7)(p11.2pter)[29.8%], +8[26.2%], del(13)(q12.3q33.1)[28.8%], t(9; 22)(q34.12; q11.23)[8.1%],
    gain(14)(q11.2qter)[27.3%], +X[29.3%] t(9; 22)(q34.12; q11.23)[10.3%]
    chr3 66500000 82500000 1.60E+07 p14.1p12.2 DEL AML del(3)(p12.2p14.1)[76.6%], del(6)(p24.1pter)[75.1%], del(6)(q14.1q14.3)[77.6%], 0
    gain(8)(q12.1qter)[72.8%]
    chr6 75500000 84500000 9.00E+06 q14.1q14.3 DEL AML del(3)(p12.2p14.1)[76.6%], del(6)(p24.1pter)[75.1%], del(6)(q14.1q14.3)[77.6%], 0
    gain(8)(q12.1qter)[72.8%]
    chr6 70000000 115000000 4.50E+07 q13q22.1 DEL AML del(6)(p22.3pter)[13.1%], del(6)(q13q22.1)[12.2%] t(15; 17)(q24.1; q21.2)[15.1%],
    t(15; 17)(q24.1; q21.2)[19.5%]
    chr6 500000 16000000 15500000 pterp22.3 DEL AML del(6)(p22.3pter)[13.1%], del(6)(q13q22.1)[12.2%] t(15; 17)(q24.1; q21.2)[15.1%],
    t(15; 17)(q24.1; q21.2)[19.5%]
    chr9 20500000 33500000 1.30E+07 p21.3p13.3 DEL AML del(9)(p13.3p21.3)[11.6%] 0
    chr18 500000 13000000 12500000 pterp11.21 DEL MDS −7[7.5%], del(18)(p11.21pter)[10.1%], del(18)(q21.2qter)[10.1%], +19[5.3%] 0
    chr18 55000000 80000000 2.50E+07 q21.2qter DEL MDS −7[7.5%], del(18)(p11.21pter)[10.1%], del(18)(q21.2qter)[10.1%], +19[5.3%] 0
    chr7 500000 159000000 158500000 pterqter DEL MDS −7[7.5%], del(18)(p11.21pter)[10.1%], del(18)(q21.2qter)[10.1%], +19[5.3%] 0
    chr5 89000000 172000000 8.30E+07 q14.3q35.1 DEL AML del(5)(q14.3q35.1)[24.4%] 0
    chrY 7000000 21000000 1.40E+07 p11.2q11.223 DEL MDS −Y[21.4%] 0
    chr9 68500000 105000000 36500000 q21.11q31.1 DEL AML del(9)(q21.11q31.1)[13.2%], −Y[15.4%] t(8; 21)(q21.3; q22.12)[26.5%],
    t(8; 21)(q21.3; q22.12)[27.9%]
    chrY 7000000 21000000 1.40E+07 p11.2q11.223 DEL AML del(9)(q21.11q31.1)[13.2%], −Y[15.4%1 t(8; 21)(q21.3; q22.12)[26.5%],
    t(8; 21)(q21.3; q22.12)[27.9%1
    chr4 134000000 139500000 5500000 q28.3q31.1 DEL MDS del(3)(q21.2q24)[83.4%], del(4)(q28.3q31.1)[65.6%], +8[82.7%] 0
    chr3 119500000 197500000 7.80E+07 q13.33qter DUP AML gain(3)(q13.33qter)[8.1%], +8[8.4%] 0
    chr10 500000 133500000 1.33E+08 pterqter DUP AML +8[27.6%], +10[29.3%] t(15; 17)(q24.1; q21.2)[34.2%],
    t(15; 17)(q24.1; q21.2)[28.8%]
    chr8 500000 145000000 144500000 pterqter DUP AML +8[27.6%], +10[29.3%] t(15; 17)(q24.1; q21.2)[34.2%],
    t(15; 17)(q24.1; q21.2)[28.8%]
    chr9 131000000 138000000 7.00E+06 q34.12qter DUP ALL gain(9)(q34.12qter)[58.2%], del(19)(p13.3pter)[53.8%], gain(22)(q11.21q11.23)[54.9%] t(9; 22)(q34.12; q11.23)[32.6%],
    t(9; 22)(q34.12; q11.23)[43.8%]
    chr22 17500000 23500000 6.00E+06 q11.21q11.23 DUP ALL gain(9)(q34.12qter)[58.2%], del(19)(p13.3pter)[53.8%], gain(22)(q11.21q11.23)[54.9%] t(9; 22)(q34.12; q11.23)[32.6%],
    t(9; 22)(q34.12; q11.23)[43.8%]
    chr11 23000000 42000000 1.90E+07 p14.3p12 DEL AML del(9)(q21.11q31.1)[87.3%], del(11)(p12p14.3)[87.6%] t(15; 17)(q24.1; q21.2)[35.0%],
    t(15; 17)(q24.1; q21.2)[32.5%]
    chr13 47000000 53500000 6500000 q14.2q14.3 DEL MDS −7[83.9%], del(13)(q14.2q14.3)[85.3%] 0
    chr12 10000000 15500000 5500000 p13.2p12.3 DEL AML del(5)(q21.1qter)[70.8%], −7[71.0%], del(12)(p12.3p13.2)[70.9%] 0
  • TABLE S4
    New SVs Identified by WGS
    Diagnosis WGS.CNA.number WGS.CNAs WGS.Recurrent.SVs
    AML
    0 0 inv(16)(q22.1p13.11)[33.3%]
    AML 1 +8[77.9%] t(9; 11)(p21.3; q23.3)[34.5%],
    t(9; 11)(p21.3; q23.3)[26.2%]
    AML 1 −X[88.4%]  t(10; 11)(p12.31; q23.3)[32.1%]
    AML 7 +2[59.0%], +4[56.4%], +6[57.7%], +8[59.0%], t(6; 11)(q27; q23.3)[37.9%],
    gain(11)(q23.3qter)[71.0%], +19[146.2%], t(6; 11)(q27; q23.3)[32.7%]
    gain(21)(q11.2qter)[60.6%]
    AML 1 +8[84.9%] t(10; 11)(p12.31; q23.3)[37.0%]
    AML 0 0 t(11; 19)(q23.3; pter)[18.0%]
    AML 0 0 t(7; 21)(p22.1; q22.12)[23.1%]
    AML 1 +8[23.6%] t(9; 11)(p21.3; q23.3)[19.7%],
    t(9; 11)(p21.3; q23.3)[20.8%]
    AML 1 gain(21)(q11.2qter)[173.8%] t(6; 11)(q27; q23.3)[28.8%]
    AML 0 0 inv(16)(q22.1p13.11)[32.5%]
    AML 3 −7[80.9%], +8[83.2%], t(9; 11)(p21.3; q23.3)[19.3%],
    del(12)(p12.2pter)[78.2%] t(9; 11)(p21.3; q23.3)[18.4%]
    AML 1 +8[74.5%] t(9; 11)(p21.3; q23.3)[32.7%],
    t(9; 11)(p21.3; q23.3)[34.0%]
    AML 0 0 t(11; 19)(q23.3; p13.11)[23.6%],
    t(11; 19)(q23.3; p13.11)[28.5%]
  • In a comparison of genetic mutations that were identified on whole genome sequencing with those that were identified on high-coverage (>500×) targeted clinical sequencing involving 102 patients, we found sensitivities of 84.6% for single-nucleotide variants and 91.5% for insertion-deletion (indel) mutations, along with a positive predictive value of more than 99% for variants with a minimum variant allele fraction of 5% (FIG. 6A). Similar performance was observed when considering only mutations in genes necessary for risk stratification in patients with AML, including a combined sensitivity of 87.5% for single-nucleotide variants and indels in ASXL1, CEBPA, FLT3, NPM1, RUNX1, and TP53 (FIGS. 16 and 17). False negatives occurred either because the variants were in subclones or were at low coverage positions on whole-genome sequencing (FIGS. 18A, 18B, 18C, 18D, 19A, and 19B); such variants were more readily detected with higher coverage sequencing (FIG. 20).
  • Clinical Feasibility and Diagnostic Yield
  • We evaluated the feasibility of using whole-genome sequencing for routine clinical testing by prospectively sequencing samples obtained from 117 consecutive patients. For this cohort, whole-genome sequencing was performed in weekly batches with a median batch size of 4 (range, 1 to 11) with the use of bone marrow aspirate samples submitted for karyotyping and FISH studies. The median total processing time was 5.1 days, which included 2 days for library preparation, 2 days for sequencing, and less than 1 day for analysis (FIG. 7A). The shortest times were about 3 days (approximately 78 hours), when clinical laboratory staffing allowed samples to be sequenced in dedicated sequencing runs immediately after library generation. Sequencing was successful in all the samples, and only 5 samples (4.3%) had less than 25× genome coverage in a single assay run. Seven samples required manual review of the automated copy-number alteration calls, with the remaining 110 samples (94.0%) needing no additional interventions to finalize the sequencing report.
  • This set of consecutive patients was also evaluated to estimate the diagnostic yield from whole-genome sequencing as compared with testing with cytogenetic analysis and targeted sequencing. This analysis was performed separately in samples obtained from patients with AML and in those obtained from patients with MDS. In the AML samples, the comparisons included clinical results from a standard FISH panel along with cytogenetic analysis and targeted sequencing to provide a realistic estimate of the expected yield of whole-genome sequencing. In this prospective cohort, results from conventional cytogenetic analysis and FISH assays in the 68 patients with AML resulted in the diagnosis of acute promyelocytic leukemia with the fusion gene PML-RARA in 5 patients and in the assignment of 27 patients to the adverse-risk group, 10 to the intermediate-risk group, and 19 to the favorable-risk group on the basis of established guidelines; 7 patients had unsuccessful or inconclusive results on cytogenetic analysis and could not be assigned to a risk group. Four patients were assigned to risk groups solely on the basis of positive FISH results for either PML-RARA (1 patient) or del(5q) (3 patients) (FIG. 7B).
  • Whole-genome sequencing that was performed in the same cohort identified new abnormalities in 17 of 68 patients (25%). These abnormalities included cryptic or complex chromosomal rearrangements in 5 patients, new copy-number alterations that resulted in a complex karyotype in 4 patients, and identification of either a normal karyotype (in 4 patients) or 1 or 2 cytogenetic abnormalities in patients with inconclusive or unsuccessful results by cytogenetic analysis (in 4 patients). Using data only from whole-genome sequencing and a PCR assay for FLT3-ITD, we reclassified 10 of 68 patients without acute promyelocytic leukemia (15%) to a risk group that differed from the one that was based on conventional testing (FIG. 21A). A similar yield was observed for the 42 prospective patients with MDS, of whom 12 (29%) had inconclusive results on cytogenetic analysis or new findings on whole-genome sequencing, and 9 (21%) were assigned to a new IPSS-R risk category, which brings the combined number of patients with a reclassified risk-group assignment to 19 of all 117 patients (16.2%) who were included in this prospective cohort.
  • Predictive Value Using Existing Genetic-Risk Categories
  • We next asked whether whole-genome sequencing could be used in place of cytogenetic analysis to predict clinical outcomes using existing genetic risk groups. To avoid the confounding effect of hematopoietic stem-cell transplantation on outcome, we focused our analysis on 71 patients with AML who did not undergo this procedure, including 41 prospective and 30 retrospective patients; 58 patients (82%) received intensive induction chemotherapy, whereas the remaining 13 were treated with hypomethylating agents. These patients were assigned to a genetic risk group on the basis of whole-genome sequencing alone or conventional testing (the combined results of cytogenetic analysis, clinical FISH results, and targeted sequencing). The FLT3-ITD mutational status based on a PCR assay was used in the two classifications.
  • Assignments that were based on conventional testing were in agreement with the results on whole-genome sequencing for 63 of 71 patients (89%); 8 patients were reassigned to a different risk category, including 5 who had new adverse-risk findings that were identified by whole-genome sequencing (FIG. 22A). Risk groups that were defined according to the two methods had the expected associations with overall survival (adjusted P=0.09 by log-rank test in groups identified by conventional testing; adjusted P=0.01 by log-rank test in groups identified by whole-genome sequencing) (FIGS. 8A and 8B). Whole-genome sequencing provided slightly better identification of patients with adverse risk and poor outcomes than conventional testing, with a hazard ratio for death of 0.32 (95% confidence interval [CI], 0.11 to 0.92) on age-adjusted Cox regression analysis, as compared with a hazard ratio of 0.66 (95% CI, 0.17 to 1.05) by conventional risk-group analysis. Similar results were observed in a larger cohort of 101 patients who were treated with either consolidation chemotherapy or stem-cell transplantation (FIGS. 23A and 24B).
  • We reasoned that whole-genome sequencing could have the greatest benefit for patients for whom cytogenetic results are unavailable at diagnosis, which occurs in up to 20% of patients with AML. Thus, we used whole-genome sequencing to evaluate 27 patients with AML who were not treated with stem cell transplantation (of whom 22 received standard induction chemotherapy), who could not be assigned to a risk group at the time of diagnosis because of unsuccessful cytogenetic analysis (in 6 patients), inconclusive results (in 13), or unknown results (in 8), and who had no reports of risk-defining events by FISH. The mean age at diagnosis in this cohort was similar to that of patients with defined cytogenetic risk (60.8 years and 54.7 years, respectively), and the median overall survival was 11.2 months (95% CI, 5.6 to 38.8) (FIG. 8C). Whole-genome sequencing analysis identified risk-defining chromosomal abnormalities in 4 patients, including KMT2A and RUNX1-RUNXT1 rearrangements in 1 patient each or a complex karyotype in 2 patients; the remaining 23 patients had either a normal karyotype or one or two abnormalities and were assigned to a risk category on the basis of mutations identified by whole-genome sequencing (FIG. 24).
  • Survival analysis of these patients showed that risk predictions that were based on whole-genome sequencing also correlated with outcomes, with significantly longer overall survival in 21 patients with intermediate or favorable risk (median survival, 20.5 months; 95% CI, 5.6 to 38.8) than in 6 patients with adverse risk (median survival, 3.3 months; 95% CI, 1.7 to 18.9; adjusted P=0.03 by log-rank test) (FIG. 8D); hazard ratio of 0.29 (95% CI, 0.09 to 0.94) by age-adjusted Cox regression analysis. This survival difference was superior to that resulting from the assignment of patients to risk groups on the basis of gene mutations alone (FIG. 25A) and was maintained when 11 additional patients with inconclusive results on cytogenetic analysis who underwent allogeneic stem-cell transplantation were included in this cohort (total of 38 patients) (FIG. 25B)
  • The above non-limiting example is provided to further illustrate the present disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples represent approaches the inventors have found function well in the practice of the present disclosure, and thus can be considered to constitute examples of modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the present disclosure.

Claims (6)

What is claimed is:
1. A computer-implemented method for the identification of clinically relevant structural variants in a subject with AML or MDS from whole genome sequencing data, the method comprising:
a. providing a whole-genome sequencing dataset, the whole-genome sequencing dataset comprising a plurality of alignments of tumor DNA sequence fragments to a reference human genome to a computing device;
b. performing, using the computing device, a structural variant analysis on the whole-genome sequencing dataset, the structural variant analysis including copy-number alteration (CNA) identification, structural variant (SV) identification, and gene-level variant identification to identify clinically relevant structural variants indicative of AML or MDS within the whole-genome sequencing dataset; and
c. producing, using the computing device, a report comprising the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis.
2. The method of claim 1, wherein copy-number alteration (CNA) identification further comprises:
a. transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of read counts over 500,000 bp nonoverlapping windows across the genome;
b. transforming, using the computing device, the plurality of read counts into a plurality of CNAs; and
c. filtering, using the computing device, plurality of CNAs to retain only CNAs greater than 5 Mbp,
3. The method of claim 1, wherein SV identification further comprises:
a. transforming, using the computing device, the alignments of the whole-genome sequencing dataset into a plurality of SV calls;
b. filtering, using the computing device, the plurality of SVs to retain only SV calls greater than 100 kbp in length; and
c. filtering, using the computing device, the SV calls greater than 100 kbp in length to identify translocations, deletions, duplications, and inversions that overlap a predefined list of recurrent and/or risk-defining SVs associated with AML or MDS.
4. The method of claim 1, wherein gene-level variant identification further comprises identifying, using the computing device, the alignments of the whole-genome sequencing dataset within about 85 kbp targeting 40 predetermined genes and gene hotspots that are recurrently mutated in AML or MDS.
5. The method of claim 1, wherein the clinically relevant CNAs, SVs, and gene-level variants identified by the structural variant analysis are indicative of a clinical outcome of the subject.
6. The method of claim 1, wherein providing the whole-genome sequencing dataset whole genome sequencing data further comprising performing whole-genome sequencing on a biological sample comprising tumor DNA from the subject with about 60× genome coverage.
US17/699,053 2021-03-18 2022-03-18 Genome sequencing as an alternative to cytogenetic analysis Pending US20220301656A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/699,053 US20220301656A1 (en) 2021-03-18 2022-03-18 Genome sequencing as an alternative to cytogenetic analysis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163162665P 2021-03-18 2021-03-18
US17/699,053 US20220301656A1 (en) 2021-03-18 2022-03-18 Genome sequencing as an alternative to cytogenetic analysis

Publications (1)

Publication Number Publication Date
US20220301656A1 true US20220301656A1 (en) 2022-09-22

Family

ID=83284039

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/699,053 Pending US20220301656A1 (en) 2021-03-18 2022-03-18 Genome sequencing as an alternative to cytogenetic analysis

Country Status (1)

Country Link
US (1) US20220301656A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113637750A (en) * 2021-07-16 2021-11-12 宁波市第一医院 Auxiliary diagnosis, prognosis diagnosis or risk stratification marker for acute myeloid leukemia and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113637750A (en) * 2021-07-16 2021-11-12 宁波市第一医院 Auxiliary diagnosis, prognosis diagnosis or risk stratification marker for acute myeloid leukemia and application thereof

Similar Documents

Publication Publication Date Title
JP7455757B2 (en) Machine learning implementation for multianalyte assay of biological samples
Chapuy et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes
Menyhárt et al. Multi-omics approaches in cancer research with applications in tumor subtyping, prognosis, and diagnosis
JP6994059B2 (en) Mutation detection and chromosomal segment ploidy
Cieślik et al. Cancer transcriptome profiling at the juncture of clinical translation
Tran et al. Cancer genomics: technology, discovery, and translation
US20180089373A1 (en) Integrated systems and methods for automated processing and analysis of biological samples, clinical information processing and clinical trial matching
US20180119137A1 (en) Integrated systems and methods for automated processing and analysis of biological samples, clinical information processing and clinical trial matching
US9447453B2 (en) Resolving genome fractions using polymorphism counts
Chakravorty et al. Gene and variant annotation for Mendelian disorders in the era of advanced sequencing technologies
US11211144B2 (en) Methods and systems for refining copy number variation in a liquid biopsy assay
Zhao et al. TruSight oncology 500: enabling comprehensive genomic profiling and biomarker reporting with targeted sequencing
Smetana et al. Chromothripsis 18 in multiple myeloma patient with rapid extramedullary relapse
US20220301656A1 (en) Genome sequencing as an alternative to cytogenetic analysis
Liang et al. Advances in multi-omics study of prognostic biomarkers of diffuse large B-cell lymphoma
Creighton Gene expression profiles in cancers and their therapeutic implications
Tang et al. Tumor mutation burden derived from small next generation sequencing targeted gene panel as an initial screening method
Khan et al. Applications of optical genome mapping in next-generation cytogenetics and genomics
Li et al. Targeted sequencing analysis of predominant histological subtypes in resected stage I invasive lung adenocarcinoma
AU2022255198A1 (en) Cell-free dna sequence data analysis method to examine nucleosome protection and chromatin accessibility
US20210202037A1 (en) Systems and methods for genomic and genetic analysis
Jones Genomics and bioinformatics in biological discovery and pharmaceutical development
US20240182981A1 (en) Identification and design of cancer therapies based on rna sequencing
Fuligni Highly Sensitive and Specific Method for Detection of Clinically Relevant Fusion Genes across Cancer
Poletti TiMMing: developing an innovative suite of bioinformatic tools to harmonize and track the origin of copy number alterations in the evolutive history of multiple myeloma

Legal Events

Date Code Title Description
AS Assignment

Owner name: WASHINGTON UNIVERSITY, MISSOURI

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DUNCAVAGE, ERIC;SPENCER, DAVID;SIGNING DATES FROM 20220321 TO 20220324;REEL/FRAME:059440/0336

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION